Skip to main content
ACS Omega logoLink to ACS Omega
. 2025 Jul 30;10(31):33999–34087. doi: 10.1021/acsomega.5c03162

From Past to Present: Gold Nanoparticles (AuNPs) in Daily LifeSynthesis Mechanisms, Influencing Factors, Characterization, Toxicity, and Emerging Applications in Biomedicine, Nanoelectronics, and Materials Science

Abhinav Sati , Suraj N Mali ‡,*, Nehal Samdani , Sivakumar Annadurai §,*, Rhuta Dongre , Nikil Satpute , Tanvi N Ranade , Amit P Pratap †,*
PMCID: PMC12355331  PMID: 40821590

Abstract

Gold nanoparticles (AuNPs) are renowned for their unique optical, electronic, and biocompatible properties, making them ideal for applications in drug delivery, diagnostics, imaging, and materials science. With green chemistry-based biological synthesis methods gaining popularity, eco-friendly alternatives to traditional chemical and physical methods techniques have emerged. Plant extract-derived AuNPs stand out for their remarkable medicinal properties, stability, and low reactivity, enhancing their biological applicability. This review explores the different synthesis methodschemical, physical, and biologicalhighlighting their advantages and challenges. We summarize the latest research (Updated until March, 2025), focusing on the most recent developments in the synthesis and multifield application of AuNPs, offering a comprehensive perspective on their potential.


graphic file with name ao5c03162_0022.jpg


graphic file with name ao5c03162_0020.jpg

1. Introduction

The current era is observing a widespread use of nanotechnology and nanomaterials. The ever-increasing demand has motivated researchers to explore and develop novel nanomaterials with enhanced properties for diverse applications. , Nanotechnology is highly versatile, with applications spanning multiple fields such as electronics, sensors, optics, mechanics, catalysis, chemistry, cosmetics, pharmaceuticals, medicine, biomedical sciences, food technology, and environmental science. Metal nanoparticles are extensively utilized across various industries due to their exceptional physicochemical properties, , including a high surface-area-to-volume ratio, enhanced catalytic activity, and tunable optical and electronic characteristics. Gold nanoparticle chemistry and physics have developed into a significant subdiscipline within the field of nanotechnology and metal nanoparticles. The unique set of optical properties of small gold particles, the size-dependent electrochemical behavior, and their remarkable chemical stability have established them as the preferred model system for investigating a variety of phenomena. Gold nanoparticles (AuNPs) are tiny gold particles, typically ranging from 1 to 100 nm synthesized through different methods such as physical, chemical, or green synthesis. They have the unique property of plasmon resonance, which enables applications in sensing, catalysis, imaging, and energy conversion. These properties are influenced by quantum effects and advanced physical processes. At the macroscopic level, gold is recognized for its distinct yellow color, chemical stability, and high redox potential. At the nanoscale, the properties of AuNPs (Figure ) arise from a blend of its electronic structure and additional effects that are associated with its extremely small size. These include a high ratio of surface atoms to bulk atoms, electromagnetic confinement due to localized plasmon resonance when interacting with optical waves, and quantum effects, which, for example, account for the transition from metallic to semiconducting behavior. , Gold nanoparticles (AuNPs) have gained significant attention in theranostic applications for cancer therapy. In the biomedical field, they are used for highly sensitive biomolecular screening, selective destruction of cancer cells via photothermal therapy, targeted protein labeling, and specific cellular drug delivery.

1.

1

Various properties of gold nanoparticles (AuNPs).

2. Synthesis of AuNPs

2.1. Physical and Chemical Methods of Synthesis

Gold nanoparticles can be synthesized through a variety of methods (Figure ). Physical methods such as micropatterning, sputtering, , pyrolysis, , laser ablation, and ball milling, , use mechanical forces or energy-based processes to generate nanoparticles (Table ). Due to the absence of any chemical moiety or process involved, the nanoparticles are high in purity along with a minimum amount of contamination. However, the imperfect surface structure of the nanoparticles and the high cost of the process, which requires a massive amount of energy to maintain high-pressure and high-temperature conditions, have particular demerits. For instance, in a study by Sylvestre et al., gold nanoparticles were synthesized using femtosecond laser ablation in aqueous solutions, hence leading to partial oxidation and a negatively charged surface. This charge, enhanced by Cl, OH, or N-propylamine, prevented the coalescence and controlled the nanoparticle size. Oxidation increased the reactivity, aiding functionalization through covalent and electrostatic interactions allowing for precise control over nanoparticle growth, producing stable, uniformly sized particles (5–8 nm) suitable for various applications in nanotechnology and materials science. In another study by Mafuné et al. AuNPs were synthesized via laser ablation of a gold plate in sodium dodecyl sulfate solution. Their absorption matched chemically prepared nanoparticles. Increased surfactant concentration reduced particle size, stabilizing them above 10–5 M. Larger nanoparticles (>5 nm) were fragmented into 1–5 nm sizes using a 532 nm laser. Similarly, Resta et al. synthesized AuNPs via pulsed laser deposition (PLD) on amorphous carbon/glass and single-crystalline MgO substrates. Despite similar nucleation rates, nanoparticle shape varied: quasi-spherical on amorphous substrates and faceted on MgO. High-energy ions (≥200 eV) enhanced nucleation, while epitaxial growth on MgO enabled shape control, following the Wulff-Kaichew theorem. Similarly, Donnelly et al. used nanosecond pulsed laser deposition (PLD) on Si and sapphire substrates in a vacuum for the synthesis of AuNPs. Atomic force microscopy confirmed nanoparticle formation below 5 nm thickness, with increasing deposition leading to coalescence. Optical absorption showed surface plasmon resonance, shifting to longer wavelengths as film thickness increased. Jankowsk et al. utilized high-voltage AC arc discharge in distilled water to synthesize AuNPs, with characterization through X-ray diffraction (XRD), transmission electron microscopy (TEM), energy dispersive X-ray spectroscopy (EDS), ultraviolet–visible (UV–vis), and inductively coupled plasma-mass spectrometry (ICP-MS) confirming nanoparticle formation, crystallinity, and stability. The synthesis process showed that increasing the discharge time resulted in larger nanoparticles, with AuNPs-2 having a crystallite size of 8.4 ± 3.2 nm and AuNPs-5 exhibiting a size of 11.6 ± 2.8 nm. This cost-effective and environmentally friendly method provides controlled nanoparticle synthesis without the need for stabilizers, making it a viable alternative to DC methods. Endla and Radhika utilized high-energy ball milling to convert microsized Au powder into nanoparticles over a 20-h process. Particle size decreases from 115 to 22 nm, with increased strain and B values. The energy of vacancy formation and Debye temperature were evaluated, showing significant effects on particle size and strain. Hatakeyama et al. synthesized AuNPs using sputter deposition in the ionic liquid, 1-butyl-3-methylimidazolium tetrafluoroborate. The size of AuNPs was influenced by target temperature, applied voltage, and capture medium temperature, while sputtering time, working distance, and discharge current, had minimal effects. Lower temperatures and higher voltages produced smaller NPs. In a study by Gromov et al., AuNPs and Au–Cu nanoalloys were synthesized using thermal evaporation, condensation, and heating in a vacuum. Due to size effects, their melting point decreased, influencing phase formation and structural deviations from bulk phase diagrams. TEM analysis revealed nanoparticle migration and fusion, affecting their final composition and stability. On the other hand, chemical-based methods (Table ) utilize the unique properties of different chemicals to mediate the synthesis of AuNPs. They are synthesized chemically by reducing gold salts like HAuCl4 using agents such as sodium citrate or NaBH4. The reduction leads to nucleation, followed by controlled growth. Stabilizing agents prevent aggregation, thus ensuring desired size and shape formation through surface interactions. Some techniques such as microemulsion, electrochemical methods, radiation-induced synthesis, , Turkevich synthesis, Brust–Schiffrin synthesis, seeding-growth technique, ascorbic acid mediated synthesis, , and synthesis with NaBH4, , have been widely used to synthesize AuNPs.

2.

2

Synthesis method for AuNPs: top-down and bottom-up approach.

1. Physical Methods for the Synthesis of Gold Nanoparticles.

Sr. No. method principle methodology parameters and conditions size & shape properties applications references
1 Pulsed Laser Ablation in Liquid (PLAL) High-energy laser pulses ablate gold in water to form NPs via plasma and condensation. 1064 nm Nd:YAG; 15 and 30 min; well-dispersed; eco-friendly 5.20 and 7.46 nm; spherical Low MIC (1.719 μg/mL S. aureus); biocompatible Antibacterial dental implants
2 Laser Ablation in Liquid (LAL) Ablation in MEM & DW using 532/1064 nm lasers to form colloids. 8.84 nm (MEM); 16.3 nm (DW); 600 μg/mL for photothermal Spherical Cytotoxic to MDA-MB-231 cells; ∼43–58% ablation Photothermal cancer therapy
3 Pulsed Laser Ablation in Liquid (PLAL) (Core–Shell Au@Fe3O4) Gold and iron oxide ablated together in liquid to form core–shell particles. 1200 mJ laser; porous silicon substrate; XRD, PL, TEM used Core–shell; 25 nm Bandgap 2.38–2.53 eV; high photodetector responsivity (0.189 A/W) Photodetectors, biosensors
4 Pulsed Laser Ablation in Liquid (PLAL) Laser ablation of gold foil in DI water; analyzed for biological response. 1064 nm, 250 mJ, 6 Hz, 300 pulses; drop-cast; FE-SEM, UV–vis, XRD Spherical; Avg. 29 nm (crystalline: 24.6 nm); SPR@550 nm Stable; Creatine Kinase levels increase with dose in rats Enzyme modulation therapy; possible drug dosage control
5 PLAL-assisted Doping Au target irradiated (500–2500 pulses) in CdS sol–gel to dope thin films. 1064 nm Nd:YAG, 700 mJ, 9 Hz; 500–2500 pulses; spin-coated films CdS: 6.6–9.3 nm (doped); grain size down to 25.8 nm Decreased bandgap (2.39 → 2.33 eV); nonlinear refraction and absorption Optical modulators, photonic devices, nonlinear optics
6 PLAL + Photoelectrochemical (PEC) Etching Au-NPs deposited on nanostructured porous Si made via photoelectrochemical etching. 400–1000 mJ; bandgap: 1.82–1.77 eV; max responsivity: 0.086 A/W at 1000 mJ Spherical; SPR at 500–580 nm Visible-NIR detection; high surface-to-volume photoresponse Quantum photodetectors; UV–vis-NIR sensors
7 PLAL (Au@CuO Core–Shell) Gold core ablated, CuO forms nanoshell in liquid; deposited on porous silicon. 1200 mJ Nd:YAG; cubic Au, monoclinic CuO; bandgap: 2.53–2.82 eV; PL + UV–vis Core–shell, spherical; Avg. 41 nm High photodetector responsivity (0.194A/W@548 nm); broad-spectrum absorption Wearable optoelectronics, photodetectors, plasmonic devices
8 PLAL (AuNP–doped CNF) Au ablated into CNF dissolved in DMF by 500–1500 pulses to tune doping. 1064 nm, 500–1500 pulses, 500–1500 mJ; CNF roughness ↓ from 158 to 106 nm; SPR @530 nm AuNPs ∼ 50.5 nm; spherical; embedded in fibrous CNF matrix Sensitivity ↑ (7.5–19.2%), recovery and response time ↑; antibacterial zone 27.10 mm (S. aureus) Gas sensors (200 °C); antibacterial coatings; flexible electronics
9 PLAL (Au@Al2O3 Core–Shell) Au plate ablated in DI water; Al placed in same jar under same laser to form core–shell NPs. 532 nm Nd:YAG, 250 pulses, 1 Hz, 9 ns; PECE PS etched with 20% HF, 20 mA/cm2, 15 min Core–shell; spherical; size increases with laser energy Bandgap 3.93–4.42 eV; max responsivity 0.084 A/W at 1000 mJ; UV–IR detection Optoelectronics, photodetectors, UV–Vis–IR sensing
10 Synthesis of Si@Au Core–Satellite Nanostructures Synthesis of Si@Au Core–Satellite Nanostructures Synthesis of Si@Au Core–Satellite Nanostructures Synthesis of Si@Au Core–Satellite Nanostructures Synthesis of Si@Au Core–Satellite Nanostructures Synthesis of Si@Au Core–Satellite Nanostructures
11 Coprecipitation + Ultrasonic Spray Pyrolysis (USP) Coprecipitation forms Fe3O4 NPs via base-induced precipitation of iron salts; USP thermally decomposes gold precursors into AuNPs that nucleate and coat the magnetic core Fe2+/Fe3+ with NH4OH yields Fe3O4; AuNPs formed on Fe3O4 via USP; separate AuNPs also observed; crystalline structure; particle size 10–200 nm; poor zeta stability (ζ ≈ 0 mV) Crystalline, mostly spherical; 10–200 nm Superparamagnetic core; reduced saturation magnetization (0.1 emu/g); poor colloidal stability; visible aggregation; AuNPs exhibit plasmonic response Biomedical imaging, drug delivery, plasmon-enhanced magnetic diagnostics
12 Coprecipitation + Ultrasonic Spray Pyrolysis (USP) for LFIA USP thermally decomposes HAuCl4 to form AuNPs in PVP-stabilized DI water; conjugated with SARS-CoV-2 S1 antigen and Rabbit IgG for diagnostic use. AuNPs (40–60 nm); 204 ppm in PBS; conjugated with 100 μg S1 antigen; borate buffer pH 8.5; incubated 30 min at 22  °C; OD change 0.91 → 0.33. Crystalline; spherical; Avg. 60  ±  20 nm Monodispersed; stable postlyophilization; effective protein binding; UV–vis confirmed conjugation; OD ↓ indicates binding strength LFIA rapid diagnostics for SARS-CoV-2; virus antigen detection; pandemic monitoring
13 Physical Sputtering + Annealing Au film sputtered onto substrates is thermally agglomerated into nanostructures via annealing, forming SERS-active surfaces with tunable morphology and enhancement properties. 10–20 mA current, 20–40 s sputtering; annealed at 300–500 °C for 1 h; substrates: silicon/quartz; cooling in air; Raman enhancement: 100–200×. Irregular Au; size and spacing increase with film thickness; roughness varies Morphology-dependent enhancement; tunable by current, time, and annealing; simple and scalable fabrication; 100–200× Raman signal boost Low-cost, scalable SERS substrates for biomedical, chemical and industrial trace analysis
14 Combinatorial Magnetron Sputtering + Annealing Au layers of graded thickness sputtered on fused silica, then annealed to form nanoparticles with spatially varying optical and sensing characteristics. Deposited Au thickness: 1.6–7 nm; annealed postdeposition; gradient profile; optical scanning and FEM modeling used to determine optimal sensing zones for Raman, gas, and molecular detection. Size varies with thickness; denser packing at 2–4 nm; optimal plasmon peak at 7 nm Tunable SERS and gas sensitivity; near-field enhancement optimized at specific thicknesses; supported by FEM modeling; high-throughput mapping Optimized SERS and molecular/gas sensing, database generation, and machine learning input for sensing material design
15 Magnetron Sputtering + Thermal Dewetting Ultrathin Au films (9–18 s sputter) deposited on glass, annealed at 500 °C for 2 h, forming tunable Au NPs with LSPR properties dependent on deposition time. Sputtering: 9–18 s; annealing: 500 °C, 2 h; RI sensitivity: 36.118–92.165 nm/RIU; optimized LOD: 1:5120 dilution for ASFV antibodies; stable and regenerable for 1 month Tunable 527–563 nm LSPR peak; NP diameter, density, spacing dependent on Au thickness High sensitivity (LOD: 5.92 ng/mL), regenerability (93% over 5 cycles), reproducibility, and long-term stability ASFV antibody biosensing, veterinary diagnostics, POF devices, medical diagnostics, food safety, environmental monitoring
16 Femtosecond Laser Ablation + White-Light Growth Two-step laser process: seed formation via femtosecond ablation in biopolymer solution, followed by white-light-induced size-controlled growth without surfactants. Laser: 800 nm, 110 fs, 1 kHz; spot: 180 μm; polymer: chitosan, PEG, etc.; NP size 2–80 nm controlled by polymer/gold ratio; dispersion ∼20% Spherical; tunable 2–80 nm; low polydispersity (∼20%) Surfactant-free, size-controlled, biopolymer-functionalized, nontoxic; stable; suitable for aqueous biosystems Biosensing via gold-dextran–concanavalin A aggregation model; biomedical and diagnostic applications
17 Laser-Induced Fragmentation (with/without Graphene Oxide) Picosecond laser-induced phase explosion fragments AuNPs; graphene oxide sheets electrostatically confine fragments, influencing diffusion, growth, and final cluster size. 1064 nm, 10 ps, 9.6 mJ, 100 kHz; pH 9, 0.3 mM NaCl; graphene oxide: 0.3 g/L; sizes: 5–20 nm (no GO) vs 40–70 nm (with GO) Spherical; with graphene oxide: larger ∼ 60 nm; without: smaller 5–20 nm Surfactant-free; graphene oxide enhances size by hindering diffusion; phase-explosion governed; size control via additives Nanocatalysis, electrochemical devices, graphene-based hybrids; platform for fragmentation studies in colloids
18 Microchip Laser Pulsed Laser Ablation in Liquid (MCL-PLAL) Laser ablation generates nanoparticles in liquid using focused high-energy pulses Gold target ablated in toluene with 10–400 mM polystyrene; 1064 nm laser, 104–550 mW, 900 ps, 100 Hz; 60 min irradiation at room temperature with stirring at 200 rpm 1.7–2.8 nm, spherical, carbon-encapsulated Size and carbon thickness tunable via laser power and polymer; strong photoluminescence with excitation-dependence Optoelectronics, photonic materials, sensing, bioimaging
19 Pulsed Laser Ablation in Liquid (PLAL) + Pulsed Laser Deposition (PLD) Laser ablation generates AuNPs in liquid; PLD forms ZrO2 films on porous Si AuNPs synthesized via PLAL; drop-cast on ZrO2 films prepared by PLD (600–1000 mJ); applied on porous Si; 3 V bias; tested for UV responsivity (350 nm) 10–15 nm, spherical Crystalline FCC Au; monoclinic ZrO2; uniform dispersion; high responsivity (0.17 A/W); QE up to 60.7% UV photodetectors, optoelectronics, high-sensitivity sensors
20 Pulsed Laser Ablation in Liquid (PLAL) Sequential laser ablation enables core–shell nanostructure formation in liquid medium Au@WO3 synthesized using Nd:YAG laser (1064 and 532 nm), 19.10 J/cm2, 1200 pulses; particle size 35–97 nm; size/shell thickness depend on laser wavelength 35–97 nm, spherical, core–shell (Au@WO3) Tunable bandgap (3.17–3.0 eV), SPR-enhanced absorption (250–550 nm), monoclinic WO3 phase, strong PL at 371–372 nm UV–Vis photodetectors, plasmonics, photocatalysis

2. Chemical Methods for the Synthesis of Gold Nanoparticles.

Sr. No. synthesis method parameters and conditions size and shape properties application suggested references
1 Turkevich method HAuCl4 (5 mM), 3 mL added to 250 mL flask with 54 mL water; Boil while stirring (400 rpm), 3 mL α-amino acid salt (20 mM) added Quasi-spherical AuNPs (several to tens of nm), irregular AuNPs (20–60 nm, for l-tyrosine, l-lysine), nanoclusters (25–95 nm, for l-(4)-hydroxyproline) Good stability, variable optical properties Therapeutics, diagnostics
2 Turkevich method with citrate Use of trisodium citrate as reducing agent, forming complex with Au3+ to Au+ species Highly stabilized AuNPs, 5–150 nm in size Stable, reliable synthesis, size variability Common in nanoparticle synthesis, diagnostics, and sensors
3 Seed-mediated growth (Turkevich method) Use preprepared citrate-stabilized Au NP seeds, controlled addition of HAuCl4, reflux at 125 °C with 500 mM sodium citrate (2.5 mM final concentration), stirring at 480 rpm Spherical, monodisperse AuNPs, size range 21–53 nm Water-dispersible, stable, uniform growth Biosensing, drug delivery, biomedical applications
4 Seed-growth method (Turkevich) 2.2 mM trisodium citrate heated to boiling, add 8.4 mM chloroauric acid, cycles of addition and heating Gold NPs of 63.9 nm for conjugation with IgY anti-N-SARS-CoV-2 Economical, reproducible, antibody conjugation SARS-CoV-2 detection, biosensors, diagnostic tools
5 Brust-Schiffrin method HAuCl4 in aqueous phase, phase-transfer agent (TOAB), reducing agent (NaBH4), alkanethiols as stabilizers Monodisperse AuNPs, size tunable between 1–5 nm Chemical stability, monodispersity, surface functionalization, stable in organic solvents Catalysis, sensing, biomedical use, nanoelectronics
6 Aqueous synthesis of tiopronin-capped gold nanoclusters Gold precursor (HAuCl4) dissolved in water, tiopronin and sodium hydroxide added for deprotonation, followed by reduction with NaBH4 Tiopronin-capped gold nanoclusters/nanoparticles, size 2.1–2.5 nm Dialysis-free synthesis, stable nucleation, photoluminescence, hydrodynamic size control Drug delivery, biomedical applications, biomolecule detection
7 Gold Nanostars (AuNSt) using PVP in DMF PVP synthesized with H2O2 or AIBN, pH 8–9, 70 °C, bases NH4OH or Na2CO3. Seed prep with citrate-capped AuNP. AuNSt (sharp tips) or spherical NPs Shape influenced by PVP impurities; H2O2 leads to sharp-tipped AuNSt, AIBN gives spherical NPs. Nanotechnology, biosensing, drug delivery.
8 Photochemical reduction of gold ions using CW & femtosecond lasers 445 nm CW laser for AuNW in HAuCl4; 800 nm femtosecond laser for AuNR, Au nanoplates Gold NPs on AuNW, Au nanoplates Hot electron-induced reduction; femtosecond laser enables precise shape control Nanophotonics, laser-assisted nanofabrication
9 Paper-based UV dosimeter via solid-state photochemical reduction of Au-complexes UV light exposure (254–365 nm), AuCl4–CTAB complex, phosphate buffer (pH 8), hydration step Gold NPs on paper UV-induced gold reduction, hydrochromic response upon hydration, color change on demand UV exposure monitoring, sterilization, UV phototherapy
10 Seed-mediated colloidal synthesis using chiral amino acids and peptides Octahedral seeds and chiral agents like l-GSH or d-GSH used. Synthesis done at room temperature in 8 h with optimized seed and peptide concentration. 432-symmetric helicoid III nanoparticles with well-defined chiral morphology. High g-factor (∼0.2), tunable CD peak, and consistent reproducibility. Chiroptical properties depend on seeds and synthesis optimization. Chiral sensing, polarization control, and biomedical applications.
11 Chemical synthesis using shape-directing agents Gold nanospheres, rods, stars, and prisms synthesized by adjusting reducing agents, surfactants, and reactants. Conditions tailored to control morphology and optical response. Nanospheres, nanorods, nanostars, and nanoprisms with controlled size and shape. Nanoprisms showed highest RIS (691.5 nm/RIU) and FoM (3.4). Sensitivity increases with particle size and is strongly shape-dependent. LSPR-based and SERS-based biosensing using shape-optimized gold nanostructures.
12 Surfactant-free colloidal synthesis at room temperature AuNPs synthesized using 0.5 mM HAuCl4, 2 mM NaOH in ethanol–water. Size controlled by shaking, stirring, or sonication. Simple, green, and reproducible approach. Quasi-spherical gold nanoparticles ranging from 5–22 nm. Smaller particles (5 nm) had highest electrochemical activity. Method enables clear observation of size effects in catalysis. Alcohol electro-oxidation, green synthesis for catalysis, size-effect studies.
13 Turkevich method using sodium citrate Reduction of NaAuCl4 with sodium citrate at ∼90 °C. Simple, rapid, and didactic synthesis with visual process steps and detailed mechanism explanation. Characterized using SEM, AFM, XRD, UV–vis, and DLS. Spherical gold nanoparticles, average size 12–25 nm. Stable AuNPs with SPR peak at 521 nm, FCC structure, good dispersity, and citrate capping. Ideal for modifying electrochemical sensors due to biocompatibility and size. Electrochemical sensors, teaching demonstrations, biomedical and plasmonic applications.
14 Seed-mediated growth using Turkevich-prepared Au seeds Presynthesized citrate-stabilized Au seeds grown by adding HAuCl4 at boiling temperature. Size controlled by adjusting precursor, seed, temperature, and reducing agent. One-step synthesis enables simplicity. Monodisperse spherical AuNPs ranging from 21–53 nm. Citrate-stabilized, water-dispersible AuNPs. Size uniformity improves with boiling. Particles suitable for functionalization with biomolecules for advanced biological applications. Drug delivery, biosensing, and bioconjugation in medicine and biology.
15 Turkevich (labile) and Ranelate-based (inert) AuNPs Turkevich: 1 mL 40 mM sodium citrate into 10 mL 1 mM HAuCl4 at boiling. Ranelate: 0.1 mL 20 mM lithium ranelate in 9.9 mL 0.202 mM HAuCl4. Both produce quasi-spherical nanoparticles; diameter not explicitly stated but suitable for SERS studies. AuNP@cit (labile) allows strong SERS via chemical/electromagnetic mechanisms; AuNP@ran (inert) suppresses SERS due to ligand rigidity, blocking electronic contact with probe molecules. SERS-based detection, understanding plasmonic interactions, chemical and electromagnetic enhancement studies.
16 Turkevich + DNA Conjugation AuNPs synthesized via Turkevich method at 92 °C with trisodium citrate, followed by DNA initiator functionalization and TdT enzyme-based enzymatic DNA elongation on surface. ∼58.3 nm, spherical, PDI < 0.2 High monodispersity, stable DNA conjugation, suitable for enzymatic synthesis platforms Enzymatic DNA synthesis, DNA data storage, DNA origami, nanorobotics, biosensing, drug delivery
17 Turkevich method (with and without PVA/PEG stabilizers) Gold nanoparticles synthesized using boiling HAuCl4 with trisodium citrate; PVA or PEG (0.03%) added to enhance stability; 20 min reflux; stored at 4 °C Bimodal size distribution; hydrodynamic diameter ∼32 nm; z-average ∼ 20 nm; PDI 0.55 PVA and PEG coatings enhanced colloidal stability and shelf life but reduced oligonucleotide sensing ability Colorimetric detection of Hepatitis C virus (HCV); visual and spectroscopic distinction of samples
18 Modified Brust–Schiffrin method using amine-functionalized liquid crystals Chloroauric acid transferred to organic phase via TOAB; reduced with LC ligand; excess LC removed via ethanol washing and centrifugation; DCM used as solvent Not explicitly stated Enhanced mesophase thermal range, Fano-like resonance in UV–vis due to 1D superstructure; improved electro-optic properties Electro-optic materials, photonic devices, plasmonic systems
19 Modified Brust–Schiffrin with ligand exchange AuNPs synthesized using TOAB as phase transfer agent; DDT as capping ligand; NaBH4 as reducing agent; purified with methanol at –20 °C; 3-azopyridine ligand exchanged postsynthesis. ∼2 nm, spherical Hydrophobic; excellent dispersibility in organic solvents; ligand photoswitching (E/Z isomerism) unaffected by surface conjugation. Photoresponsive materials; reversible nematic-to-isotropic transitions in liquid crystalline hosts under UV/green light irradiation.
20 Citrate reduction + ligand exchange (TDG); MPA-based conjugation (EDCl/NHS, DCC/DMAP) AuNPs synthesized using 300 mg HAuCl4·3H2O, 157 μL MPA in 150 mL methanol; NaBH4 (6–12 mmol, 30 mL) added dropwise. TDG: 0.2% w/v in DMSO; conjugation via esterification with 100 mg DG, 85 mg DCC, 20 mg DMAP. Purified by centrifugation at 13,000–14,500 rpm. MPAm1:27.8 nm (spherical), MPAm2:58.8 nm (polycrystalline); MPAm1-DG: ∼ 53 nm, ζ-potential –30 mV SP-ICP-MS confirmed narrow size distribution; 27.6% loading; DG conjugates showed improved cancer cell selectivity and reduced toxicity vs TDG; >90% synthetic yield Targeted prostate cancer therapy; safer diosgenin delivery; advanced nanomedicine with selective cytotoxicity to DU-145 cells

2.2. Green Synthesis of AuNPs

Apart from the traditional physical and chemical methods of synthesis, green synthesis (Table ) has gained a lot of attention primarily due to its essence of sustainability and low toxicity. There are several eco-friendly and biocompatible techniques to synthesize AuNPs that utilize plant extracts, bacteria, and microbes. The chemical composition and concentration of reducing agents in organic extracts are significant and can vary, which in turn influences the characteristics of the final product. These variations lead to differences in size and shape, which in turn affect the function and application of the material. Many biomolecules including, phenols, flavonoids, amino acids, proteins, enzymes, amines, aldehydes, ketones, carboxylic acids, and alkaloids act as electron donors, which enable the reduction of cationic gold to form AuNPs. The properties of the resulting nanoparticles are determined by factors such as the plant extract concentration, the type of metal salt used, the pH of the reaction mixture, and the reaction temperature. ,

3. Green Synthesis of AuNPs: Plant type, Biomolecules, Reaction Conditions, Shape and Size, and Applications.

Sr. No. plant/source used plant part used biomolecules responsible gold salt used reaction conditions size & shape of AuNPs method characterization techniques applications references
1 Ziziphus spina-christi Leaves Alkaloids, phenols, flavonoids, proteins, vitamins HAuCl4·3H2O (Gold Chloride) 70 °C, 60 min, aqueous extract 31.26–58.06 nm, spherical Green synthesis using aqueous plant extract under controlled temperature and time, ensuring nanoparticle formation with stability and bioactivity for therapeutic applications. TEM, FE-SEM, AFM, XRD Effective in photothermal therapy for breast cancer treatment, leveraging near-infrared radiation to induce localized hyperthermia and apoptosis in cancer cells.
2 Cordia myxa L. Leaves Flavonoid glycosides, phenolic derivatives HAuCl4·3H2O (Gold Chloride) 80 °C, 90 min, aqueous extract 56.49–89.38 nm, spherical Green synthesis involving biomolecule-assisted reduction of gold salts, yielding nanoparticles with controlled morphology and enhanced biocompatibility for medical applications. TEM, FE-SEM, AFM, XRD Utilized in targeted cancer therapy by combining gold nanoparticles with near-infrared irradiation to enhance cytotoxicity and induce apoptosis in breast cancer cells.
3 Capsicum annum Fruit (Dried) Phenols, flavonoids, capsaicin, secondary metabolites HAuCl4 (Gold Chloride) 90 °C, rapid mixing, aqueous extract 20–30 nm, spherical Green synthesis via aqueous extract of dried fruit powder, acting as both reducing and stabilizing agent to form highly stable gold nanoparticles with biomedical relevance. TEM, FT-IR, EDAX, XRD, ζ-potential Demonstrates strong antioxidant, anti-inflammatory, and antiangiogenic properties, making it useful in cancer treatment by inhibiting blood vessel formation and oxidative damage.
4 Rutin extract Flavonoid (Rutin) Rutin (Flavonoid) HAuCl4 (Gold Chloride) 532 nm laser, 150 J/cm2, 300 s exposure 40 nm, spherical Green synthesis using Rutin as a stabilizing and reducing agent, forming plasmonic nanoparticles suitable for photothermal applications. TEM, UV–vis, MTT assay Effective in plasmonic photothermal therapy (PPTT) for breast cancer, with high selectivity for MCF-7 cancer cells and minimal cytotoxicity to normal cells. Potential for antibody-functionalized targeted therapy.
5 Mentha spicata L. Essential Oil Monoterpenes (Menthol, Carvone, Limonene) HAuCl4 (Gold Chloride) Room temperature, 24 h reaction time 19.61 nm, spherical Green synthesis using essential oil as a reducing and stabilizing agent at room temperature, ensuring eco-friendly nanoparticle formation with enhanced bioactivity. TEM, DLS, FT-IR, XRD, UV–vis, MTT assay Exhibits antibacterial, antioxidant, and cytotoxic properties, with potent free radical scavenging ability and selective cytotoxicity against HEPG-2 liver cancer cells. Suitable for biomedical and pharmaceutical applications.
6 Chitosan & Reduced Graphene Oxide (rGO) Chitosan (Different MW) Chitosan (Polysaccharide) HAuCl4 (Gold Chloride) Varying MW of chitosan, room temperature 9.29–13.03 nm, spherical, immobilized on rGO Green synthesis using chitosan as a reducing and stabilizing agent, preventing AuNP aggregation and enhancing catalytic efficiency. FT-IR, XRD, XPS, SEM, FESEM, EDS, TEM, HRTEM, TGA Highly efficient and stable catalyst for the reduction of 4-nitrophenol (4-NP) in the presence of NaBH4. Potential application in catalysis for reducing aromatic nitro compounds.
7 Halymenia pseudofloresii (Red Algae) Whole Algae (Dried) Phenolic compounds, carboxyl groups HAuCl4 (Gold Chloride) 60 °C, 20 min, aqueous extract 27 nm, cubic and rectangular Green synthesis using seaweed extract as a reducing and capping agent, ensuring bioactive-stabilized nanoparticle formation. UV–vis, XRD, FTIR, SEM Exhibits strong antioxidant, antibacterial, and anticancer activities, effectively inhibiting Staphylococcus aureus, Lactobacillus, and Pseudomonas aeruginosa. Shows potent cytotoxicity against A549 lung cancer cells (IC50 = 19.02 μg/mL) and LN-18 glioblastoma cells (IC50 = 32.46 μg/mL). Induces apoptosis via ROS generation, making it a potential anticancer agent.
8 Ziziphus zizyphus Leaves Phenolic compounds, flavonoids, alkaloids HAuCl4·3H2O (Gold Chloride) Boiling extraction, 5 min 50 nm, spherical Green synthesis of gold nanoparticles using Ziziphus zizyphus leaf extract, followed by purification via dialysis and column separation. SEM, TEM, DLS, ζ-potential, ICP-MS, Histopathology Evaluated for acute and chronic toxicity in albino mice. Gold accumulation was highest in the spleen, liver, and kidneys. Mild to moderate histopathological changes were observed in kidney tissue, with minimal effects on heart and blood glucose levels. Suggests need for further toxicity assessments for biomedical applications.
9 White Pitaya (Hylocereus undatus) Extract (Pulp) Phenolic compounds, polysaccharides HAuCl4 Magnetic stirring (30 min) + heating (65 °C, 2 min) 18.0 ± 0.7 nm, spherical Green synthesis via the reduction of gold ions using bioactive compounds in H. undatus extract under controlled thermal and magnetic stirring conditions, leading to the formation of stable AuNPs. UV–vis Spectroscopy, TEM Electrochemical sensor for hydroxychloroquine detection, pharmaceutical analysis, clinical diagnostics, biosensing, environmental monitoring, and biomedical applications
10 Andrographis paniculata Leaves Polyphenols, flavonoids, triterpenoids HAuCl4 1.5 mL extract, 0.5 mM HAuCl4, 60 °C, 30 min, pH 6 10–15 nm, spherical Green synthesis through the bioreduction of Au3+ to Au0 by polyphenolic and flavonoid compounds in A. paniculata extract under optimized reaction temperature and pH, ensuring uniform nanoparticle dispersion. UV–vis Spectroscopy, TEM, FTIR, XRD, EDX, DLS, ζ-potential, SEM Lead ion detection, dye degradation (methylene blue, methyl orange, crystal violet), antibacterial (B. subtilis, Salmonella enterica, S. aureus, Escherichia coli), antioxidant activity, biosensing, wastewater treatment
11 Glaucium flavum Leaves Alkaloids (glaucine, aporphine, protoberberine, protopine), polyphenols HAuCl4 1 mL extract +1 mL 10 mM HAuCl4, stirred for 1 h 32 nm, spherical Green synthesis utilizing alkaloid-rich G. flavum extract as a reducing and stabilizing agent, forming monodisperse AuNPs through a controlled nucleation and growth mechanism. UV–vis Spectroscopy, TEM, DLS, SEM, XRD, FTIR, ζ-potential Antiviral (H1N1 virus), antioxidant, biosensing, drug delivery, biocompatibility studies, biomedical applications
12 Ephedra Whole plant extract Alkaloids (ephedrine, pseudoephedrine), polyphenols, tannins, flavonoids, glycosides HAuCl4·3H2O 4 mL extract +6 mL 10–3 M HAuCl4, stirred at 25 °C for 3 h, pH 4 1.3–15.6 nm, spherical and triangular Green synthesis through the reduction of gold salt by alkaloids and polyphenols in Ephedra extract at ambient temperature, promoting nanoparticle stabilization via capping biomolecules. UV–vis Spectroscopy, FTIR, TEM, HRTEM, XRD, EDX, XPS, ζ-potential Antioxidant, antipyretic (reduced fever by 83.3%), antiasthmatic, antibacterial (S. aureus, B. subtilis, L. monocytogenes, E. coli , P. aeruginosa, S. typhimurium), antifungal (Candida albicans, A. nigra, A. flavus), biosensing, drug delivery
13 Oroxylum indicum Plant extract Flavonoids HAuCl4 Microwave-assisted synthesis, 3 min at 800 W 5.25 ± 1.00 nm, spherical Microwave-assisted green synthesis utilizing O. indicum extract, where flavonoids facilitate the rapid reduction of gold ions under electromagnetic radiation, enhancing nanoparticle formation kinetics. UV–vis Spectroscopy, FTIR, XRD, HRTEM, EDX, XPS, ζ-potential Antibacterial (S. aureus, E. coli), catalytic reduction of 4-nitrophenol, potential biomedical applications
14 Satureja khuzestanica Aerial parts Phenolic compounds, flavonoids, terpenoids HAuCl4 Extract and gold salt mixed, ultrasonic treatment (40 min, 60 °C), followed by rotary evaporation 20–30 nm, cubic Green synthesis employing ultrasonic-assisted reduction, where bioactive phytochemicals in S. khuzestanica extract act as reducing and capping agents, ensuring controlled morphology and stability of AuNPs. UV–vis Spectroscopy, SEM, XRD, FTIR Antiparasitic (hydatid cyst protoscoleces), apoptosis induction via caspase-3 activation, ultrastructural cell damage, low cytotoxicity toward normal human cells, potential biomedical and antiparasitic applications
15 Prunus persica (Peach) Pulp extract Phenolic compounds (chlorogenic acid, catechins, epicatechins, anthocyanins), polyphenol oxidase enzyme HAuCl4 1 mL peach extract +0.25 mL 6.0 mM HAuCl4, stirred at 500 rpm for 3 min at 25 °C 11.5 ± 1.5 nm, spherical Green synthesis via the bioreduction of Au3+ using phenolic compounds and polyphenol oxidase enzyme in P. persica extract. The synthesized AuNPs were further incorporated into graphene to enhance sensor conductivity. UV–vis Spectroscopy, ATR-FTIR, TEM, SEM, EIS, Cyclic Voltammetry Electrochemical sensor for butylated hydroxyanisole (BHA) detection in food matrices, biosensing, environmental analysis, and antioxidant screening
16 Moringa oleifera + Titanium dioxide (TiO2) Leaf extract Flavonoids, polyphenols, alkaloids HAuCl4·3H2O Extract heated (100 °C, 1 h), cooled, filtered; Gold salt added to TiO2 (1:1:1), stirred (60 °C, 1 h), then calcined (200 °C, 2 h) 10 ± 5 nm, uniformly dispersed Green synthesis of TiO2@Au nanocomposite using M. oleifera extract as a reducing and stabilizing agent. The synthesized AuNPs were uniformly deposited onto TiO2 for enhanced photocatalytic activity. UV–vis Spectroscopy, FTIR, Raman, XRD, XPS, TEM Photocatalytic degradation of ciprofloxacin under visible light, environmental remediation, wastewater treatment, phytotoxicity studies with Pistia stratiotes
17 Thyme (Thymus kotschyanus) Extract Polyphenolic compounds HAuCl4 Green synthesis using plant extract, room temperature 20–25 nm, spherical Green synthesis using Thymus kotschyanus extract as a natural reducing and stabilizing agent, ensuring controlled nucleation, preventing aggregation, and forming monodisperse AuNPs without harsh chemicals under mild reaction conditions. UV–Vis, FT-IR, XRD, TEM Used as catalysts in the reduction of 4-nitrophenol to 4-aminophenol, a crucial step in wastewater treatment. Catalysts were recyclable for six cycles with over 90% efficiency.
18 Green tea (Camellia sinensis) Leaves Polyphenols, flavonoids HAuCl4 Green synthesis using aqueous extract, room temperature Spherical, stable Green synthesis using Camellia sinensis extract as a biocompatible reducing agent, avoiding toxic precursors, ensuring high stability, and enabling large-scale production under mild conditions. UV–Vis, SEM, Optical fiber analysis Functionalized for biosensing of CD44 cancer biomarker, significantly enhancing sensitivity in optical fiber biosensors, achieving ultralow detection limits (0.111 pM), and enabling real-time cancer biomarker detection.
19 Chitosan Biopolymer Chitosan (reducing and capping agent) HAuCl4 Heating for 15 min in 1% acetic acid 18 ± 4 nm (AuNPs), 25 ± 5 nm (6MP-AuNPs), spherical Green synthesis using chitosan as a reducing and stabilizing agent, ensuring high stability, biocompatibility, and eco-friendly nanoparticle production. 6MP drug was loaded via sonication. UV–Vis, ζ-potential, SEM Developed as a hybrid nanocomposite (6MP-AuNPs) for combinatorial chemo-photothermal therapy. Demonstrated enhanced anticancer efficacy with laser irradiation, reducing cell viability by 63% at 1.25 μM.
20 Garlic (Allium sativum) Cloves Sulfur compounds, organosulfur compounds HAuCl4 Boiling in water, reaction at 95 °C, color change to wine red within 15 min 7–21 nm, spherical Green synthesis using Allium sativum extract as a reducing agent, enabling biocompatible, eco-friendly synthesis with high yield and stability. UV–Vis, HRTEM, FESEM, EDX Exhibited strong antifungal activity against Candida species (C. albicans, C. krusei, C. tropicalis, C. guilliermondii). MIC80 values ranged from 40 to 0.31 μg/mL, with antifungal mechanisms involving ROS generation, membrane disruption, and metabolic inhibition. Showed low toxicity on human cells, highlighting potential for biomedical applications.
21 Marine soft coral (Sarcophyton crassocaule) Whole coral extract Organic compounds (polysaccharides, proteins, polyphenols) HAuCl4·3H2O Reaction at 40–45 °C, color change to ruby red at 540 nm, completed in 30 min 5–50 nm, spherical and oval First-time green synthesis using Sarcophyton crassocaule extract, ensuring biofabrication under mild reaction conditions with high stability. UV–vis, TEM, SEM, FT-IR, ζ-potential, TGA Exhibited strong antibacterial (V. cholerae, S. aureus), antioxidant (DPPH: 85%, RP: 82%), and antidiabetic (α-amylase inhibition: 68%, α-glucosidase inhibition: 79%) properties. Also functioned as a catalytic agent for the degradation of 4-NP, MR, and BPB, showing 91% catalytic efficiency. Potential for therapeutic applications against type II diabetes and as an eco-friendly material for pollutant degradation.
22 Marine algae (Solieria tenuis) Whole algae extract Phycoerythrin (PE) HAuCl4·3H2O Incubation at 4 °C for 48 h, controlled PE-to-gold ratios 10–25 nm, avg. Sixteen nm, spherical Green synthesis using Solieria tenuis extract to mediate the formation of Au@PE NPs, leveraging PE as both reducing and capping agents to enhance solubility and catalytic activity. UV–vis, Fluorescence spectroscopy, FT-IR, TEM, XPS, XRD Efficient catalyst for the reduction of 4-nitrophenol (4-NP) to 4-aminophenol (4-AP) and degradation of cationic dyes (MB, Safranine O) in wastewater. Demonstrated high catalytic efficiency (k298K = 0.36× 10 2 s –1 ), offering an eco-friendly solution for environmental remediation.
23 Amphipterygium adstringens extract Bark Polyphenols, flavonoids, tannins HAuCl4 Shock wave-assisted and ultrasound-assisted extraction using aqueous, ethanolic, and methanolic extracts 10 nm (maceration), 50–100 nm (shock wave, quasi-spherical), 100–150 nm (ultrasound, irregular/triangular) Green synthesis using A. adstringens extracts obtained through shock wave and ultrasound-assisted extraction UV–vis, XRD, FTIR, TEM, ζ-potential Cytotoxic activity against leukemia cells (Jurkat), IC50: 87 μM (ultrasound-assisted AuNPs), 94.7 μM (shock wave-assisted AuNPs); potential as drug nanocarriers for cancer therapy
24 Azadirachta indica extract Leaf Polyphenols, flavonoids HAuCl4 Reduction using aqueous leaf extract 73.74–171.45 nm, crystallite size 5.68–13.25 nm, alloy and core–shell structures Green synthesis of Au-based bimetallic nanoparticles (BNPs) UV–vis (SPR: 423–557 nm), XRD, FTIR, ζ-potential (−17.96 to −33.11 mV) Nanocatalysts for textile dye degradation (Rh–B and MO), recyclable and reusable
25 Elaeis guineensisJacq. (Oil Palm) Leaf Extract Flavonoids, Phenols HAuCl4 0.69 mM HAuCl4, Room Temp 10–30 nm, Spherical Green synthesis using E. guineensis extract as a reducing agent UV–Vis, FT-IR, TEM, XRD, ζ-potential, Raman Spectroscopy Used in SERS for enhanced Raman intensity and sensitive molecular detection.
26 Coleus scutellarioides (L.) Benth Leaf Extract Phenolic Compounds HAuCl4 1 mM HAuCl4, 25 °C, Stirring 3 min 40.10 nm, Spherical Green synthesis using C. scutellarioides extract as a bioreductant and stabilizing agent UV–Vis, XRD, TEM, DLS, FT-IR, FESEM, EDX Exhibits significant anticancer activity against MDA-MB-231 cells and antioxidant properties.
27 Allium cepa (Red Onion Peel) Peel Extract Flavonoids, Polyphenols HAuCl4·3H2O 75 mM HAuCl4, Room Temp 6.08–54.20 nm, Spherical Green synthesis using A. cepa peel extract as a bioreductant UV–vis, XRD, FT-IR, TEM, EDX, TGA-DTA, AFM Displays antibacterial, antifungal, antioxidant, and anticholinesterase activities for Alzheimer’s therapy.
28 Pseudomonas aeruginosa Bacterial Culture Proteins, Enzymes HAuCl4 pH 9.0, 24 h at 58 °C Small, Spherical, Less Aggregated Microbial green synthesis using P. aeruginosa for optimized redox potential ATR-FT-IR, TEM, UV–vis Shows high antitumor efficacy against PC3 prostate cancer cells, with reduced IL-6 levels.
29 Artemisia annua Hairy Root Extract Hydroxybenzoic Acids, Flavonoids, Caffeic Acid HAuCl4 Ethanolic (70%) extract, controlled conditions 15–40 nm, Triangular and Spherical Green synthesis using A. annua extract as a reducing agent HPLC, MALDI-MS, TEM, UV–vis Exhibits strong photocatalytic activity for Methylene Blue degradation and potential biomedical applications.
30 Halimeda macroloba (Green Macroalgae) Whole Extract Bioactive Compounds HAuCl4 Sunlight exposure, natural bioreduction 18.72 nm, Spherical Green synthesis using H. macroloba extract for efficient nanoparticle production UV–vis, FT-IR, TEM, XRD, SEM-EDX, XPS, PSD High photocatalytic degradation (97.23% MB, 89.91% MO) for industrial dye removal under sunlight.
31 Citrus sinensis (Orange Peel) Peel Extract Polyphenols, Flavonoids HAuCl4 1 mM HAuCl4, 100 °C, 30 min 37 nm, Spherical Green synthesis using C. sinensis peel extract as a reducing agent UV–vis, XRD, SEM, FT-IR, TEM Development of a highly sensitive, noninvasive electrochemical glucose sensor for diabetes monitoring in saliva.
32 Peganum harmala Leaf Extract Hydroxyl, Carboxyl, Polyphenolic Groups HAuCl4 1 mM HAuCl4, optimal ratio 4:1, pH 14, 12 h stirring 12.6–35.7 nm, Oval and Spherical Green synthesis using P. harmala leaf extract as a reducing and stabilizing agent UV–vis, FT-IR, SEM, EDX Significant antibacterial activity against E. coli (25 mm) and antifungal efficacy against Candida albicans (31 mm).
33 Passiflora ligularis (Sweet Granadilla) Pulp Extract Polyphenols, Antioxidants HAuCl4·3H2O 1 × 10–3 M HAuCl4, pH 6, 25 °C, 6 h 8.43–13 nm, Spherical and Triangular Green synthesis using P. ligularis extract for controlled nanoparticle formation UV–vis, XRD, HRTEM, EDX, ζ-potential Strong neuroprotective effects against autism, reducing neuroinflammation, oxidative stress, and apoptosis in brain tissue.
34 Citrus sinensis (Orange Peel) Peel Extract Polyphenols, Hydroxyl Groups HAuCl4 0.165 mM HAuCl4, 100 °C, 1 h Spherical, Stable in Water Green synthesis using C. sinensis extract as a reducing and capping agent UV–vis, FT-IR, DLS, SEM Selective colorimetric detection of Pb2+ ions in water with LOD of 0.05 μM, applicable for real water samples.
35 Equisetum diffusum (Horsetail) Leaf Extract Polar Bioactive Compounds HAuCl4·3H2O 1.26 mM HAuCl4, Sunlight exposure (4–5 min), 30 °C Cubic, Well-Dispersed Green synthesis using E. diffusum extract under sunlight irradiation for nanoparticle formation UV–vis, FT-IR, SEM-EDX, PXRD, DLS, ζ-potential Highly selective Pb2+ colorimetric sensor (LOD 4.4 μM), antimicrobial activity, and 96.5% photocatalytic degradation of methylene blue.
36 Curcumin and Doxorubicin-Loaded AuNPs - Trisodium Citrate, Curcumin, Doxorubicin HAuCl4 2.0 mM HAuCl4, 19.4 mM trisodium citrate, 0.2 mM drug Spherical, Stable Green synthesis using anticancer drug-loaded gold nanoparticles UV–vis, DLS, TEM, Hyperspectral Microscopy, MTT Assay Ultrasound-triggered drug release for targeted cancer therapy, effective intracellular uptake, and enhanced cytotoxicity.
37 Curcuma longa (Turmeric Root) Root Extract Curcuminoids, Polyphenols HAuCl4 1% turmeric extract, 1–5 mM HAuCl4, 24 h in dark 8.5 nm, Spherical Green synthesis using C. longa extract for controlled nanoparticle formation UV–vis, FT-IR, TEM, XRD Sensing of Al3+ and Cr3+ ions (LOD: 0.83 and 1.19 nM), catalytic reduction of 4-NP, degradation of MB and MO dyes in wastewater.
38 Lilium longiflorum (Easter Lily) Stem Extract Organic Compounds HAuCl4·3H2O 1 mM HAuCl4, 25 °C, 6 mL extract 4.97 nm, Spherical Green synthesis using L. longiflorum extract as reducing and capping agent UV–vis, FT-IR, TEM, SEM, EDX, HRTEM High catalytic efficiency in 4-NP and MB reduction (rate constants 1.50 min–1 and 1.29 min–1), promising for environmental remediation.
39 Aconitum violaceum Jacquem. ex Stapf Crude Extract Alkaloids, Flavonoids HAuCl4 Stirring for 50 min, pH 9, Centrifugation (6000 rpm for 15 min) Spherical, Triangular, <100 nm Green synthesis using A. violaceum extract as a reducing and capping agent for stable AuNPs formation under controlled pH conditions. SEM, TEM, EDX, XRD, UV–vis, FTIR, XPS, ζ-potential Effective antibacterial agent against L. acidophilus and E. coli, strong antioxidant, and efficient photocatalyst for methylene blue degradation under UV light.
40 Annona muricata Leaf Extract Phytoconstituents HAuCl4 Agitation at room temperature until deep purple color appears, incubation for 24 h Not specified Green synthesis using A. muricata leaf extract as a reducing and capping agent for stable AuNP formation under ambient conditions. UV–vis, TEM, SEM, XRD Exhibits significant anticancer activity against SCC-15 cells by inducing apoptosis via upregulation of Bax and p53 genes and downregulation of Bcl-2.
41 Acorus calamus Leaf Extract Bioactive Compounds HAuCl4·3H2O 5 mL extract mixed with 95 mL of 1 mM gold salt, stored at room temperature for 30 min until ruby red color appears Hexagonal, Face-Centered Cubic, 30–50 nm Green synthesis using A. calamus leaf extract as a reducing and stabilizing agent for controlled AuNP fabrication with optimized reaction parameters. XRD, UV–vis, FTIR, TEM Demonstrates neuroprotective potential by inhibiting cholinesterase activity, crossing the blood-brain barrier, and reducing oxidative stress for potential Alzheimer’s treatment.
42 Catha edulis (Khat) Leaf Extract Phytochemicals HAuCl4 0.75 mL extract added to 30 mL of 1 mM boiling HAuCl4, stirred for 45 min, color change to wine red, centrifuged after 24 h Spherical, 17.3 ± 3.7 nm Green synthesis using Khat leaf extract as a reducing and capping agent for AuNP formation under controlled conditions. UV–vis, TEM Developed a colorimetric nanosensor for BPA detection in plastic bottled water with high sensitivity, low detection limit (0.09 ng/mL), and recovery of 86.7–98.0%.
43 Quercetin (Flavonoid) - Quercetin HAuCl4 Quercetin solution (0.05%) mixed with water, gold salt added, stirred at RT until color change from yellow to red/purple Spherical, ∼10 nm Green synthesis using quercetin as a reducing agent to form Au/TiO2 hybrid nanocomposites with controlled gold loading UV–vis, TEM, ζ-potential, Band Gap Energy Measurement Enhanced photocatalytic degradation of ciprofloxacin antibiotic under visible and UV light, reducing TiO2 band gap from 3.05 to 2.85 eV, achieving up to 95% degradation efficiency.
44 Diospyros kaki L. (Persimmon) Leaf Phytochemicals from leaf extract HAuCl4·3H2O 0.1 and 0.5 mL extract added to 0.5 mM HAuCl4·3H2O at 25 °C, continuous stirring, microwave extraction 25.99 nm (XRD), 45.44 ± 1.095 nm (Zetasizer), spherical/elliptical Green synthesis using aqueous leaf extract UV–vis, XRD, TEM, FTIR, Zetasizer Biomedical, catalysis, and environmental applications
45 Ginkgo biloba Leaf Flavonoids, terpenoids, polyphenols HAuCl4 1:1 ratio with 1 mM HAuCl4, pH 5, 50 °C, magnetic stirring (500 rpm) 18 ± 4 nm, monodispersed, crystalline Green synthesis using aqueous leaf extract FTIR, UV–vis, XRD, TEM, ζ-potential Anticancer therapy via apoptosis induction in nasopharyngeal carcinoma cells
46 Synechococcus moorigangae strain InaCC M208 (Marine Microalga) Whole cell (Ethanol extract) Phytoconstituents (Flavonoids, proteins, polysaccharides) HAuCl4 1:1 ratio with 1 mM HAuCl4, 37 °C, 22 h incubation 8.2 nm, spherical, SPR at 552 nm, ζ-potential – 21.5 mV Green synthesis using ethanol extract UV–vis, TEM, XRD, FTIR, PSA, EDX Antibacterial and antioxidant properties for nanomedicine applications
47 Spirulina platensis (Cyanobacterium) Whole cell (Aqueous extract) OH-, carbonyl, amino acids AgNO3 1 mL extract mixed with 5 mL of 1 mM AgNO3, exposed to sunlight for 10 min, incubated in dark at 30 °C for 24 h 50–70 nm, spherical Green synthesis using S. platensis extract UV–vis, SEM, FTIR Antibacterial, antioxidant, and biocompatibility for biomedical applications
48 Scutellaria barbata D. Don (Scu) Whole plant (Aqueous extract) Reducing sugars, flavonoids, polyphenols HAuCl4 12 mL of 2 mM HAuCl4 mixed with 2 mL of extract, reaction at 60 °C for 30 min 50 nm, spherical Green synthesis using Scu extract as reducing and stabilizing agent TEM, DLS, XRD, XPS, UV–vis Photothermal and chemo-photothermal therapy for cancer treatment, enhancing apoptosis and necrosis in tumor cells
49 Tangerine (Citrus reticulata) Peel Extract (TPE) Peel Phytochemicals (Polyphenols, Flavonoids, Terpenoids) HAuCl4 10 mM HAuCl4 in 10 mL water, mixed with 10 mL TPE extract, reaction at 25 °C for 60 min 26 ± 5 nm, spherical Green synthesis using TPE as reducing and capping agent UV–vis, XRD, FTIR, EDX, FESEM, TEM Antibacterial, antifungal, antioxidant (92.7% DPPH scavenging), photocatalytic degradation of dyes (Methyl orange & Rhodamine B)
50 Plum (Prunus domestica) Peel Extract (PPE) Peel Phenolic compounds (Hydroxyl, Carboxyl, Polyphenols) HAuCl4·3H2O 5 mL of 1 mM HAuCl4 mixed with 30 mL of DES (Betaine-Urea & Choline chloride-Urea), heated at 65 °C for 45 min 5–12 nm, spherical Green synthesis using DES-based PPE as reducing and stabilizing agent UV–vis, FTIR, SEM, XRD, EDX, ζ-potential Antioxidant (ABTS IC50 = 110 mg/mL), skincare (moisturizing cream with 0.5–2% AuNPs), antibacterial (E. coli, B. subtilis)
51 Cucurbita moschata (Pumpkin) Fruit Peel Extract (CM) Peel Phytochemicals (Polyphenols, Flavonoids, Carboxyl & Hydroxyl groups) HAuCl4·3H2O 1:3 ratio of 250 mL of 10 mM HAuCl4 solution and 750 mL plant extract, stirred at 50 °C for 15 min at 750 rpm 18.10 nm, spherical, cubic crystal structure Green synthesis using CM peel extract as reducing and stabilizing agent UV–vis, FTIR, TEM, AFM, SEM, EDX, TGA-DTA, XRD, ζ-potential Antimicrobial (S. aureus, E. coli, C. albicans at 0.004–0.64 μg/mL), Anticancer (IC50 at 50 μg/mL for cancer cell lines Sk-Ov-3, CaCo2, A549)
52 Strobilanthes kunthiana Leaf extract Polyphenols, flavonoids, alkaloids HAuCl4 pH 7, 70 °C, 1 h reaction time 31 nm, spherical Green synthesis using Strobilanthes kunthiana leaf extract as a bioreducing and stabilizing agent for eco-friendly gold nanoparticle production. UV–vis, FTIR, XRD, FE-SEM, TEM, DLS, EDX Used in dopamine sensing, electrochemical biosensors, neurochemical detection, and medical diagnostics for precise and selective neurotransmitter quantification.
53 Halodule uninervis (HUE) Ethanolic extract Phytochemicals (hydroxyl, carbonyl groups) HAuCl4·3H2O 70–80 °C, 30 min sonication, color change to dark purple, centrifugation, lyophilization Small, spherical, <50 nm, ZP Green synthesis using H. uninervis extract as a reducing and stabilizing agent for eco-friendly and cost-effective gold nanoparticle production. UV–vis, SEM, DLS, XRD, TGA, FTIR Biogenic AuNPs exhibit anticancer properties via apoptosis induction, making them potential candidates for targeted cancer therapy and drug delivery.
54 Punica granatum (Pomegranate) Juice Polyphenols (punicalagin, ellagic acid, flavonoids), carbohydrates HAuCl4 Room temperature, 4 h stirring (120 rpm), centrifugation, solvent washing Nanostructured AuNPs, 20–50 nm, tunable characteristics, ZP Green synthesis using pomegranate juice as an effective reducing and stabilizing agent, minimizing environmental impact and utilizing agri-food waste. UV–vis, MALDI FT-ICR MS, LC-ESI-MS/MS Biogenic AuNPs show potential in food packaging and dermatology, with antimicrobial and antioxidant properties aiding in shelf life extension and skincare formulations.
55 Anemopsis californica Isopropanoic extract Bioactive phytochemicals aiding in nanoparticle stabilization HAuCl4 24 h agitation with graphene-based material, maintained as colloidal solution or vacuum filtered Homogeneous distribution of AuNPs on graphene-based material, ∼10–40 nm, ZP Green synthesis using A. californica extract for in situ nanoparticle formation and integration with graphene for enhanced nanocomposite properties. UV–vis, TEM, SEM-EDS, XPS, Raman Graphene-based AuNPs nanocomposites enhance SERS for chemical sensing, biomedical diagnostics, and environmental monitoring, offering improved sensitivity and detection limits.
56 Punica granatum (Pomegranate) Juice Carbon dots (CDs) from polyphenols and sugars HAuCl4 180 °C, 6 h hydrothermal treatment, followed by gold salt reduction using NaBH4 AuNPs-modified carbon dots (CDs), 2–10 nm, ZP Green synthesis of CDs from pomegranate juice followed by AuNPs decoration to create optical neurotransmitter nanosensors for dopamine detection. UV–vis, SEM, TEM, FL AuNPs-CDs sensor efficiently detects dopamine in biological samples with high sensitivity and selectivity, enabling advanced diagnostics for neurological disorders.
57 Madhuca indica Flower extract Phytochemicals acting as reducing and stabilizing agents HAuCl4 Rapid green synthesis, UV absorbance at 550 nm Spherical, 20.34 ± 4.36 nm, ZP Green synthesis of AuNPs using Madhuca indica flower extract, providing a cost-effective and eco-friendly approach for nanomaterial synthesis. UV–vis, XRD, FTIR, TEM, FE-SEM, DLS, EDX MI-AuNPs exhibit strong anticancer effects by targeting cancer stemness and EMT in HNSCC, demonstrating therapeutic potential with minimal toxicity.
58 Arctium lappa Flowers Quercetin (Flavonoid) None (Laser Ablation) Soxhlet extraction, hydrolysis, crystallization, laser ablation Spherical AuNPs 80–160 nm, AgNPs 20–40 nm, ZP Green synthesis of AuNPs and AgNPs using quercetin extracted from A. lappa, ensuring eco-friendly nanoparticle production. UV–vis, HPLC, FTIR, NMR, SEM, TEM, XRD AuNPs and AgNPs exhibit high antioxidant activity, with AgNPs showing superior free radical scavenging, highlighting their potential for antioxidant therapy and biomedical applications.
59 Psidium araca (Sour Guava) Peel extract Polyphenols (Phenolic acids, flavonoids) HAuCl4·3H2O 50 °C, 1 h stirring, response surface optimization, NaBH4 addition Gold nanorods (AuNRs), ∼30 nm, LSPR 700–800 nm Green synthesis using Psidium araca peel extract as a weak reducing agent for gold nanorods biosynthesis, replacing ascorbic acid. UV–vis, TEM, LSPR Optimized synthesis of anisotropic AuNRs for photothermal therapy, targeted drug delivery, and diagnostic imaging in biomedical applications
60 Sphaeranthus amaranthoides Leaf extract Phenolic compounds Chloroauric acid (HAuCl4) Sunlight exposure for 2 h, PVA stabilization Anisotropic nanoparticles Green synthesis using Sphaeranthus amaranthoides leaf extract as a natural reducing agent under solar irradiation SPR, HRTEM, ζ-potential analysis Biocompatible gold nanoparticles for targeted drug delivery, exhibiting cytotoxicity against THP-1 Acute Monocytic Leukemia cells
61 Sodium lignosulfonate Biopolymer Lignosulfonate Chloroauric acid (HAuCl4) Ultrasonic irradiation at 60 °C for 30 min 20–30 nm, spherical Green synthesis using sodium lignosulfonate under ultrasonic conditions for gold nanoparticle stabilization and reduction UV–vis, FT-IR, FE-SEM, TEM, EDX, ICP-AES Catalysis in Suzuki–Miyaura coupling, antigastric cancer agent via PI3K-Akt-mTOR modulation, high biocompatibility for biomedical applications
62 Croton draco Branch extract Polyphenols, flavonoids Chloroauric acid (HAuCl4) Different extraction methods, 25 ± 2 °C, UAE method preferred 10–50 nm, spherical Green synthesis using Croton draco aqueous extracts with high antioxidant capacity for reducing and stabilizing AuNPs UV–vis, TGA, DLS, SEM, XRD, FT-IR Excellent antibacterial (99% Gram-positive, 87% Gram-negative) and anti-inflammatory (97%) properties, potential biomedical applications
63 Stachytarpheta jamaicensis Whole plant extract Polyphenols, flavonoids Chloroauric acid (HAuCl4) Room temperature, plant extract-mediated reduction and stabilization 60.3 nm, spherical Green synthesis using Stachytarpheta jamaicensis extract for eco-friendly nanoparticle formation with therapeutic potential UV–vis, FT-IR, SEM, PSA Strong anticancer activity in MCF7 breast cancer cells (IC50 = 19.53 μg/mL), downregulation of MYC and CCND1 oncogenes, potential in nanomedicine
64 Scrophularia striata Boiss Flower and stem extract Polyphenols, flavonoids Silver nitrate (AgNO3) Ultrasonication for 15 min at room temperature, optimized plant extract stored at 4 °C 56.1 nm (stem), 62.5 nm (flower), spherical In-situ green synthesis of Ag NPs on polyamide (PA) fabric using plant extract as a reducing and stabilizing agent UV–vis, DLS, SEM, XRD Enhanced dyeing performance (K/S increase of 64–100%), excellent antibacterial activity (100% inhibition against E. coli and S. aureus), retained 88–79% activity after 10 washes, potential for medical and hygienic textile applications
65 Tangerine peel extract Peel extract Flavonoids, polyphenols Chloroauric acid (HAuCl4) 40 °C, 15 mM gold concentration, 60 min 26 ± 5 nm, spherical Green synthesis using Tangerine peel extract (TPE) as a reducing and capping agent UV–vis, XRD, FT-IR, EDAX, FE-SEM, TEM Strong antibacterial (MIC: 31.25–62.5 μg/mL), antifungal (Candida albicans), 92.7% antioxidant (DPPH assay),6effective photocatalytic degradation (88.6–93.2% UV, 67.3–74.1% sunlight)
66 Croton Caudatus Geisel Leaf extract Polyphenols, antioxidants Chloroauric acid (HAuCl4), Hexachloroplatinic acid (H2PtCl6·6H2O) 60 °C, reaction time 30 min 12–33 nm, rectangular Single-step green synthesis of Au–Pt bimetallic nanoparticles without external capping or stabilizing agents UV–vis, FT-IR, PXRD, TEM, EDAX, SAED Strong antibacterial, antifungal, and anticancer effects (IC50 = 37.17 μg/mL in HeLa cells), potential for bimetallic nanoparticle drug development
67 Euphorbia acaulis Leaf extract Polyphenols, flavonoids Silver nitrate (AgNO3), Chloroauric acid (HAuCl4) Room temperature, optimized leaf extract-mediated reduction 1–100 nm, spherical Green synthesis of silver (SNPs) and gold nanoparticles (GNPs) using E. acaulis as a natural reducing and capping agent UV–vis, HR-SEM, EDX, TEM, AFM, XRD, FTIR Anticancer (MCF-7, SNP IC50: 59.87 μg/mL, GNP IC50: 91.074 μg/mL), apoptosis and DNA damage induction, superior antibacterial (E. coli 17 mm, S. aureus 10.77 mm), larvicidal (Aedes aegypti, SNP LC50: 20.81 mg/L, GNP LC50: 51.10 mg/L), photocatalytic methylene blue degradation

For instance, Srinath et al. reported the biosynthesis of biocompatible gold nanoparticles (AuNPs) using Brevibacillus formosus isolated from the Hutti gold mine, India. The bacterial metabolites acted as reducing and stabilizing agents. Synthesized AuNPs (5–12 nm, spherical) were characterized using UV–vis, Fourier transform infrared (FTIR), dynamic light scattering (DLS), and TEM and also exhibited strong antibacterial activity against S. aureus along with biocompatibility with chicken RBCs, highlighting their potential for biomedical applications. In another study by Singh and Kundu, biosynthesis of gold nanoparticles (AuNPs) using Pseudomonas aeruginosa and Rhodopseudomonas capsulata was carried out. The pH significantly influenced size and shape, with spherical 10–20 nm AuNPs at pH 7 and nanoplates at pH 4. Altaf et al. reported the green synthesis of gold nanoparticles (AuNPs) using Iris kashmiriana rhizome extract with spherical AuNPs (∼80 nm). These nanoparticles exhibited strong antibacterial, antibiofilm, and antiadherence properties, particularly against Streptococcus mutans, making them effective for coating orthodontic appliances. Daramola et al. evaluated the bacteria-mediated synthesis of gold nanoparticles (AuNPs) using Bacillus subtilis and P. aeruginosa. The process involved the microbial reduction of gold salts, producing stable, biocompatible nanoparticles of size 118 nm. Abu-Elghait et al. developed optimal conditions for the biosynthesis of gold (AuNPs) using Trichoderma saturnisporum. The ideal conditions for AuNPs were pH 6.94, 33.2 °C, and 1.21 mmol, offering a more efficient alternative to traditional synthesis methods. Another study, Omole et al. explored the microbial synthesis of gold nanoparticles (AuNPs) using Lysinibacillus fusiformis and HAuCl4 solution. The synthesized AuNPs were characterized by UV–vis, SEM, EDX, DLS, TEM, XRD, and FT-IR analyses, revealing spherical particles with a mean size of 121.2 nm. These AuNPs demonstrated significant antibacterial activity against multidrug-resistant bacteria from chronic wounds, indicating their potential for pharmaceutical applications in treating infections caused by Multi-Drug Resistant (MDR) bacteria. Table effectively summarizes all advantages and disadvantages of physical, chemical and various green synthesis methods in synthesis of AuNPs.

4. Comparative Overview of Physical, Chemical, and Green Synthesis Methods for Gold Nanoparticles (AuNPs) .

method type synthesis method principle advantages disadvantages cost scalability purity issues biocompatibility limitations references
Physical Laser Ablation A pulsed laser is focused on a bulk gold target submerged in liquid, causing vaporization and nanoparticle formation via plasma-induced cavitation, without chemical reagents. High purity, no chemicals, clean process Expensive, low yield, not scalable High Low Very low Very High Poor scalability, costly equipment, difficult size control
Ball Milling Mechanical grinding of bulk gold using high-energy balls in a rotating chamber produces nanoparticles by collision and shear forces, typically in inert conditions. Simple, scalable, solvent-free Contamination from media, broad size distribution Low High Moderate Moderate Particle aggregation, contamination from grinding media
Evaporation–Condensation Gold is evaporated in a high-temperature zone and condensed into nanoparticles in a colder inert atmosphere, avoiding chemical reagents or solvents. High purity, solvent-free Needs vacuum, poor control High Low Low High Needs inert gas, limited shape control
Ultrasonic Spray Pyrolysis Ultrasonic waves generate aerosol droplets from gold precursor solutions; these undergo pyrolysis in a high-temperature furnace to yield gold nanoparticles. Continuous, scalable, uniform synthesis Requires high T, complex setup Moderate High Moderate Moderate–High Agglomeration risk, byproduct formation
Sputter Deposition Gold atoms are ejected from a gold target in vacuum using energetic ions and deposited onto a substrate or dispersed in a liquid medium. High purity, precise control Slow process, expensive setup High Low Very low High Limited for large-scale production
Microwave Irradiation Microwave energy rapidly heats gold precursor solutions via dielectric heating, inducing nucleation and growth of gold nanoparticles without conventional thermal conduction. Fast, uniform heating, simple Size variability, depends on solvent Low–Moderate Moderate Moderate High (in aqueous setup) Uneven nucleation, less suitable for scale-up
Arc Discharge High-voltage electric arc vaporizes gold electrodes in a gas environment, with subsequent cooling and condensation forming gold nanoparticles. High temperature, no reagents Broad size, low yield, safety issues Moderate Low Low–Moderate High Risk of spark ignition, broad distribution
Thermal Decomposition (Physical) Solid gold compounds are thermally decomposed at elevated temperatures to form gold nanoparticles without solvents, often under inert or reducing atmospheres. Simple, solvent-free Needs high T, low shape control Moderate Moderate Low High Not suitable for heat-sensitive applications
Chemical Turkevich Method (Citrate) Gold salt (e.g., HAuCl4) is reduced in aqueous medium using trisodium citrate, which also stabilizes the formed nanoparticles via surface adsorption. Easy, aqueous, good control Surface-bound citrate may interfere Low High Moderate High (after washing) Citrate residues may hinder functionalization
Brust-Schiffrin Method A two-phase system uses NaBH4 to reduce Au3+ in the presence of alkanethiols, forming stable organic-phase monolayer-protected nanoparticles. Stable, tunable size Toxic organics and thiols Moderate Moderate Moderate–High Moderate Limited biocompatibility without purification
Seed-Mediated Growth Small gold seeds are used to control the anisotropic or isotropic growth of larger nanoparticles using additional precursors and surfactants. Excellent size/shape control Multistep, impurity-sensitive Moderate High Moderate High Requires precise conditions and surfactant use
Microemulsion Nanoscale water droplets within surfactant-stabilized micelles act as confined reactors for the reduction of gold salts to form nanoparticles. Narrow size distribution Surfactant removal is difficult Moderate–High Low–Moderate Moderate Moderate–High Thermodynamically unstable systems
Sonochemical (Chemical) Acoustic cavitation from ultrasonic waves generates radicals and extreme conditions that drive the reduction of gold ions into nanoparticles. Fast, fine dispersion Requires optimization, broad distribution Low–Moderate Moderate Moderate Moderate–High Cavitation effects difficult to control
Electrochemical Reduction Gold ions in solution are reduced to metallic nanoparticles at the cathode under applied potential in an electrochemical cell. High purity, no chemicals Electrode degradation, low throughput Low–Moderate Low–Moderate Very low High Limited industrial viability
Photochemical Reduction Light-sensitive compounds generate reducing agents upon UV or visible irradiation, which then reduce Au3+ to Au0 nanoparticles in solution. Mild, tunable via light Needs photoactive reagents Moderate Low–Moderate Moderate High Requires UV/visible light control
Radiolytic Reduction High-energy radiation (e.g., γ-rays) induces solvent radiolysis, generating radicals that reduce gold ions to form ultrapure nanoparticles. High purity, no reducing agents Requires radiation source, expensive High Low Very low High Inaccessible in many laboratories, safety concerns
Polyol Method Gold salt is reduced by polyols (e.g., ethylene glycol) at high temperature, which also act as stabilizers to control growth. Good size control, no extra reductants Organic solvent removal required Low–Moderate Moderate Moderate Moderate Viscous solvent complicates purification
Alcohol Reduction Alcohols like ethanol or methanol reduce gold salts upon heating, with temperature and alcohol concentration influencing nucleation and growth. Simple, low cost Needs heating, larger size distribution Low Moderate Low–Moderate High Temperature-sensitive reaction
Chemical Vapor Deposition Gold precursor vapor is decomposed or reacts on a heated substrate to form nanoparticles or thin films under controlled atmosphere. Pure, controllable High vacuum, substrate limited High Low Very low High Industrial-scale usage limited to thin films
Sol–Gel Method Gold precursors are hydrolyzed and condensed to form gels, which on drying and calcination yield gold nanoparticles embedded in matrix. Morphology control, embedding capability Long processing time Moderate Moderate Moderate Moderate–High Solvent residues, drying/cracking during gelation
Green Plant Extract-Mediated Synthesis Phytochemicals like flavonoids and polyphenols in plant extracts reduce gold ions and stabilize nanoparticles without toxic chemicals. Eco-friendly, abundant raw materials, high biocompatibility Batch-to-batch variability, poor size/shape control Very Low Moderate Low–Moderate Very High Reproducibility issues, extract complexity
Microbial Synthesis (Bacteria) Bacterial enzymes or metabolites mediate reduction of Au3+ ions to Au0, with intracellular or extracellular nanoparticle formation. Sustainable, extracellular synthesis possible, biosafe Requires sterile growth, slow process, difficult purification Low Low–Moderate Moderate High Contamination risk, low yields
Fungal-Mediated Synthesis Fungal species secrete redox-active biomolecules that facilitate gold ion reduction and nanoparticle stabilization, often extracellularly. Higher yield than bacteria, extracellular synthesis Long incubation, complex downstream processing Low Low–Moderate Moderate High Optimization of culture conditions needed
Algal-Mediated Synthesis Algae contain reducing metabolites (e.g., polysaccharides, proteins) capable of converting gold ions into nanoparticles under mild conditions. Eco-friendly, renewable, low cost Less explored, slow kinetics Low Low Low–Moderate High Lower efficiency, extraction of active compounds needed
Enzyme-Mediated Synthesis Purified enzymes catalyze the bioreduction of Au3+ to Au0 under aqueous and controlled conditions, yielding pure and uniform nanoparticles. High selectivity, clean mechanism Enzyme purification, high cost Moderate Low Very low Very High Enzyme stability and reusability
Polysaccharide-Assisted Synthesis Natural polysaccharides (e.g., starch, cellulose) reduce and stabilize AuNPs via hydroxyl or aldehyde functional groups in water. Biodegradable, safe, stable dispersion Some polysaccharides may remain bound Low Moderate Low–Moderate Very High Surface passivation may hinder activity
Amino Acid/Protein-Based Synthesis Amino acids or proteins like BSA act as reducing and stabilizing agents under mild conditions to form biocompatible nanoparticles. Mild conditions, excellent biocompatibility Size distribution varies, protein denaturation risk Low Moderate Moderate Very High Functional group interference, protein cost
Honey-Mediated Synthesis Natural sugars and enzymes in honey serve as reducing agents for gold ions, forming capped nanoparticles with inherent antimicrobial properties. Natural, accessible, inherently antimicrobial Viscosity interferes with uniform synthesis Low Moderate Moderate High Consistency issues due to honey composition variability
Natural Gum/Resin-Assisted Synthesis Natural gums or resins contain polysaccharides and polyphenols that facilitate reduction and stabilize AuNPs in aqueous systems. Eco-safe, good stabilizing properties May alter NP surface chemistry Low Moderate Low–Moderate Very High Surface capping may affect functionalization
Biopolymer-Assisted Synthesis Biopolymers like chitosan or gelatin reduce and stabilize gold nanoparticles through functional group interactions under aqueous or mild conditions. Biodegradable, low toxicity, stable nanoparticles May interfere with surface functionality Low Moderate Low–Moderate Very High Batch inconsistency, polymer–metal interactions complex
a

Comments based on available literature.

2.3. Biosynthetic Mechanism of AuNPs

The biosynthesis of gold nanoparticles (AuNPs) typically follows a straightforward two-step process that does not require extreme temperature or pressure conditions. Initially, a biological extractsuch as one derived from plants, bacteria, or fungiis combined with a solution of chloroauric acid (HAuCl4). This interaction leads to the reduction of gold ions (Au3+) to elemental gold atoms (Au0). In the second phase, the nucleated gold atoms undergo growth and stabilization, resulting in the formation of AuNPs, as illustrated in Figure .

3.

3

Schematic representation of AuNPs biosynthesis mechanism and mechanism of the formation and stabilization of gold nanoparticles by polyphenolic compounds. Adapted with permission from ref . Copyright 2022, MDPI, Basel, Switzerland.

The synthesis is often visually confirmed by a distinct color change in the solution, indicating nanoparticle formation. The chemical reduction of Au3+ to Au0 in the presence of water can be represented by the reactions depicted in above Figure .

2.3.1. Reducing Potential of Plant Phytochemicals

Phytochemicals present in plant extractssuch as amino acids, proteins, carbohydrates, phenolic acids, flavonoids, and terpenoidsplay crucial roles in the green synthesis of metal and metal oxide nanoparticles. These bioactive compounds act as natural reducing and stabilizing agents, enabling nanoparticle formation under mild, eco-friendly conditions without the need for toxic chemicals or harsh physical processes. The literature suggests that the extent of synthesis of AuNPs is directly linked with reduction potential of plant extract. Nevertheless, these plant extracts have varieties of phytomolecules of different classes such as polyphenols, terpenes, etc. It has been seen that there are 3 key factors influencing the efficiency of this biosynthesis process: (1) the degree to which metal ions are reduced by compounds in the extract, (2) the concentration of reducing agents, and (3) the composition of bioactive compounds that stabilize the resulting AuNPs. It is very likely that higher the amount of content of reducing substances, higher would be rate of formation of AuNPs and promotes the creation of smaller nanoparticles, and enhances their stability. − , Amino acids and proteins reduce metal ions and stabilize nanoparticles through electron donation and capping, while carbohydrates (e.g., polysaccharides) contribute to particle size and morphology control due to their hydrophilic and catalytic properties. Phenolic acids and flavonoids facilitate metal ion reduction via hydrogen or electron transfer, often through redox or keto–enol transitions. Huang et al. elaborated in details mechanisms of phenolic acids in biosynthesis of AuNPs (Scheme ). One of such representative mechanism is illustrated below, which was proposed by Manyuan and Danwanichakul. In concern work, they illustrated synthesis of AuNPs with the help of spent coffee ground extract.

1. Proposed Reactions Involved in the Formation of AuNPs from HAuCl4 via Phenolic Acids.

1

Terpenoids, commonly found in essential oils, also assist in nanoparticle synthesis, particularly silver nanoparticles apart from AuNPs, due to their strong reducing capacity. Hossanisaadi et al. reviewed studies assessing the ability of plant extracts to reduce gold ions. The review examined extracts from 27 plant speciesincluding Rosa damascena, Juglans regia, Caccinia macranthera, etc.many of which are traditionally used in Middle Eastern medicine. Extracts were prepared from different plant parts, and the study identified 28 additional plant species with effective gold ion-reducing capabilities. These extracts successfully facilitated gold nanoparticle synthesis. , Additionally, the success of metal nanoparticle (MNP) biosynthesis depends on the electrochemical potential of the specific metal ion involved. Noble metal salts have reduction potentials ranging from 0.35 to 1.0 V, and metal ions can be reduced to nanoparticles if the extract’s reduction potential exceeds +0.16 V. , Table gives an idea about the reducing potentials of various phytochemicals reported for the synthesis of AuNPs.

5. Reducing Potentials of Plant Phytochemicals Reported in the Literature .
plants compounds/phytochemicals particle size (nm) shape references
Parkia roxburghii Proteins 5–25 Spherical
Murraya koenigii Polyphenols, flavonoids, alkaloids 20 Spherical, triangle
Mentha longifolia Phenolic flavonoids, tannins, etc. 10.23 ± 2 Oval
Anacardium occidentale Proteins, polyols, gallic acid, tannins 17 Spherical
Prunus serrulata Phenolic compounds, proteins 20–50 Hexagonal
Zingiber officinale Ascorbic acid, oxalic acid 5–20 Spherical, irregular (hexagonal, triangular)
Indigofera tinctoria Polyphenols, flavonoids, etc. 6–29 Spherical, hexagonal, triangular
Bauhinia purpurea Polyphenols 20–100 Triangular, hexagonal, nanorods
Cibotium barometz Flavonoids, phenolic acids, fatty acids 5–20 Spherical
Jasminum sambac Polyphenols, flavonoids, terpenoids 20 Spherical
Coleus forskohlii (1) Phenolic compounds 10–30 Spherical
Coleus forskohlii (2) Forskolin, proteins 15–35 Hexagonal
Memecylon edule Saponin 10–45 Triangular, circular, hexagonal
Mussaenda glabrata Alkaloids, tannins, flavonoids, steroids 10.59 Spherical
Stereospermum chelonoides Polyphenolic compound (lignans) 27.19 ± 5.96 Spherical
Rivea hypocrateriformis Polyphenols 20–30 Spherical
Gloriosa superba Glycosides, tannins 20–50 Triangular, spherical
Glycyrrhiza uralensis Flavonoids, polyphenols, glycyrrhizin 10–15 Spherical
Aerva lanata Polyphenols, flavonoids, alkaloids, etc. 10–30 Spherical, hexagonal, triangular plate
Dendropanax morbifera Polysaccharides 10–20 Polygonal, hexagonal
Angelica pubescens Maxim Flavonoids, phenols, sesquiterpenes 10–30 Spherical, icosahedral
Amorphophallus paeoniifolius Phenolic compounds (quercetin) 13.3 Spherical, polygonal
Asparagus racemosus Phenolics, flavonoids, etc. 10–50 Spherical
Chrysopogon zizanioides Alkaloids, phytosterols 123–138 Cubic
Memecylon umbellatum Saponins, phenolics, proteins, quinones 15–25 Spherical, triangular, hexagonal
Platycodon grandiflorum Triterpenoidal platycodon saponin 14–15 Spherical (major), triangular (minor)  
Pulicaria undulata Phenolic compounds (quercetin, kaempferol, etc.) 5–12 Regular (spherical), irregular (triangular, hexagonal, plates)
Rhodiola rosea Flavonoids, polyphenols, terpenoids, etc. 12–18 Irregular shape
Garcinia mangostana Flavonoids, phenolics, carbohydrates, glycosides 15.37–44.20 Spherical
Stemona tuberosa Lour Not specified 20–30 Irregular shape
Actinidia deliciosa Proteins 7–20 Spherical
Rosa canina L. Phenolic compounds 26 Quasi-spherical
Aspalathus linearis Polyphenols, aspalathin 7.5 ± 0.34 Hydra-like
Ficus retusa Phenolic compounds 10–25 Spherical
Mukia maderaspatana Flavonols (quercetin, phloroglucinol) 20–50 Spherical, triangular, circular
Clerodendrum inerme Phenolics, flavonoids, etc. 5.82 Spherical
Trapa natans var. bispinosa Roxb. Phenolic compounds (gallic acid, quinones) 25 ± 2 Spherical
Panax ginseng Meyer Ginsenosides, polyphenols, reducing sugars, acidic polysaccharides 5–10 Spherical
Eleutherococcus senticosus Phenolic compounds, reducing sugars, proteins 189 Face-centered cubical
Cornus mas Polyphenolic compounds 5–30 Pseudospherical
a

Adapted and modified with permission from ref . Copyright 2023, MDPI.

2.3.2. Reducing Potential of Microbial Enzymes

The biosynthesis by these microorganisms can occur via two distinct pathways: extracellular and intracellular, depending on where the nanoparticle formation takes place (Figure ). In extracellular biosynthesis, metal ions are reduced outside the bacterial cell through enzymatic activity, leading to nanoparticle formation in the surrounding medium. In contrast, intracellular biosynthesis involves the uptake of metal ions into the cell, where enzymatic processes reduce them internally to form nanoparticles ,

4.

4

Schematic representation of AuNPs biosynthesis mechanism through microorganisms. Adapted from ref . Copyright 2022, MDPI, Basel, Switzerland.

Gholami-Shabani and colleagues developed a cell-free method for synthesis of AuNPs using α-NADPH-dependent sulfite reductase purified from E. coli. An average sized AuNPs of 10 nm, were confirmed from this novel route. In another review, Shedbalkar et al. covered detailed aspects of mechanisms of microbial AuNPs synthesis. Physicochemical parameters like temperature, pH, and substrate concentration influence intracellular AuNP synthesis and morphology, with monodispersity achievable through their optimizationthough comprehensive studies are lacking. Proteins and amino acids such as cysteine, tyrosine, and tryptophan play key roles in AuNP biosynthesis and stabilization. Free amino or cysteine groups bind AuNPs for stabilization, while tyrosine and tryptophan contribute to NP formation and reduction at high pH through specific binding and redox activity. Protein type also affects AuNP capping and stability. Several other review articles have also been published detailing recent advances in microbe assisted AuNP synthesis. , Chaurasia and colleagues beautifully summarized list of various microbial sources known for producing extracellular and intracellular enzymes involved in AuNPs synthesis along with their influence of particle sizes and shapes. (Table )

6. Overview of Key Microbial Sources Known for Producing Extracellular and Intracellular Enzymes Involved in Gold Nanoparticle Synthesis .
Sr. No. microbial source shape size (nm)
1 Coprinus comatus   <100
2 Cladosporium sp. Spherical 5–10
3 Bacillus subtilis   20–25
4 Pleurotus ostreatus Uneven spherical 10–30
5 Trichoderma harzianum Spherical 32–44
6 Rhizopus oryzae Flower-like 43 ± 19
7 Cladosporium oxysporum AJP03 Quasi-spherical 72.32 ± 21.80
8 Magnusiomyces ingens LH-F1 Spherical, plates (triangle, hexagon, etc.), irregular 80.1 ± 9.8
9 Acinetobacter Spherical, triangular, polyhedral 19
10 Klebsiella pneumonia Spherical 10–15
11 Ureibacillus thermosphaericus 50–70
12 Shewanella oneidensis Round 12
13 Neurospora crassa Round 20–50
14 Brevibacterium casei 10–50
15 Yarrowia lipolytica Triangles 15
16 E. coli Hexagonal, triangular 20–30
17 Rhodopseudomonas capsulata Round 10–20
18 Pseudomonas aeruginosa 15–30
19 Sargassum wightii Planar 8–12
20 Plectonema boryanum Cubic <10–25
21 Fusarium oxysporum (Au–Ag alloy) Round 8–14
22 Rhodococcus sp. Round 8–12
a

Adapted and reproduced with permission from ref Copyright 2022, Elsevier.

Table explains various factors affecting synthesis of AuNPs.

7. Factors Affecting the Synthesis of AuNPs .
factor details
pH Affects NP shape, size, and biomolecule interaction. Acidic conditions (pH 2.5–5) promote faster aggregation and growth of medium to large NPs; basic pH (7–9) slows growth.
Temperature Influences nucleation rate, particle growth, and monodispersity. Higher temperatures yield smaller, more uniform NPs. Green synthesis methods operate below 100 °C; physical methods may exceed 350 °C. Below 50 °C can cause polydispersity and slow reactions.
Pressure Affects NP morphology and growth rate. Ambient pressure settings favor biological reduction processes and lead to more efficient NP synthesis.
Time Reaction duration influences NP size, aggregation, and completion of gold ion reduction. Example: Full reduction achieved after 60 min in palm leaf-based synthesis, beyond which wavelength stabilizes. Prolonged time may cause aggregation or degrade stability.
Concentration of Plant Extract/Biomass Higher concentrations typically lead to enhanced NP formation and morphological variations. Optimal extract levels must be established for consistent size and shape.
Salt Concentration Regulates NP yield and characteristics. Increased salt (e.g., 0.7 mM) enhances synthesis efficiency, but excessive salt may destabilize particles.
Reducing and Stabilizing Agent Concentration Agents like trisodium citrate influence reduction rate and NP stability. Higher citrate levels lead to smaller, more uniform NPs due to better control over nucleation and stabilization. The adsorption rate of the stabilizer is key in determining NP size and preventing aggregation.
Synthesis Method Chemical (e.g., citrate reduction), physical, or green synthesis methods each have distinct requirements for temperature, pH, and reactants. Method selection impacts reproducibility, particle size control, and environmental impact.
a

Well explained in the ref .

2.4. Effect of Different Parameters on the Appearance and Functionality of Gold Nanoparticles

A wide number of factors such as size, shape, and type of environment are responsible for the interaction of the AuNPs with light. A coordinated oscillation of electron charge, which is in resonance with the frequency of visible light, is generated by the interaction of free electrons with the oscillating electric fields of a light ray traveling near a colloidal nanoparticle. The size or form of the nanoparticles can be changed to modify the surface plasmon resonance, creating particles with customized optical properties for various purposes. The unique interaction with light, surface plasmon resonance, changes depending on particle sizes. The smaller ones appear red, while larger ones reflect bluish or purplish hues. Shape plays a key role in how cells absorb these particles, with triangular and rod-shaped nanoparticles being taken up more efficiently than star-shaped ones. While larger particles tend to circulate longer and serve as better delivery agents, smaller ones can be more toxic. Yue et al. showed that gold nanoparticle size and shape affect siRNA delivery. Larger particles (50 nm spheres, 40 nm stars) had higher cellular uptake and escaped endosomes, unlike smaller 13 nm spheres. This highlights the importance of nanoparticle design in enhancing functionality for effective gene delivery applications. Xie et al. evaluated how shape influences gold nanoparticle uptake in RAW264.7 cells. Star, rod, and triangle-shaped nanoparticles showed varying internalization, with triangles achieving the highest uptake. Different shapes engaged distinct endocytosis pathways, demonstrating that nanoparticle geometry significantly affects cellular uptake and can guide effective drug delivery system design. These characteristics make gold nanoparticles highly adaptable for medical applications such as imaging, therapy, and drug delivery. Figure gives an idea about various kinds of Au nanostructures.

5.

5

Transmission electron microscope (TEM) images for various kinds of Au nanostructures (a) nanospheres, (b) nanodisks, (c) nanorods, and (d) cubic nanocages. Reproduced from ref . Copyright 2021, with permission of Elsevier.

2.5. Characterization of AuNPs

The characterization of AuNPs is usually done via modern analytical techniques mentioned in Figure .

6.

6

Various Characterization techniques used for AuNPs.

2.4.1. UV–Visible Absorption Spectroscopy

UV visible absorption spectroscopy is usually performed to study the optical properties and band gap of nanoparticles. When a PerkinElmer 2 spectrometer was used for characterization, it showed a peak in range of 300–900 nm via capturing surface plasmon resonance (SPR) feature for gold nanoparticles. It has been observed that AuNPs shows a strong absorption peak around 520 nm due to SPR and it remains the same irrespective of change in particle shape or the surrounding material. Similarly, on application of gold nanoparticle on APS (aminopropylsilane) -treated glass, we can see the SPR peak shifting to a higher wavelength (red shift) followed by the band broadening due to the electromagnetic interactions between nearby particles. , This is one of the most helpful techniques to find the size, concentration, and aggregation level of gold NPs. While operating, spectrum registration may take some time and the extinct spectra of AuNPs can be derived via Mie theory. Furthermore, in one case, an Electrochemiluminescence biosensor based on gold nanoparticles was synthesized and on characterization, it was observed that a 5 nm gold nanoparticle showed a peak at 515 and 20 nm at 535 nm (Figure A,B). This demonstrated the size dependent optical property because of the localized SPR (LSPR). The 20 nm one was more effective in increasing the electrochemiluminescence signals leading to a better electron transfer. Iqbal et.al, synthesized sodium alginate (SA) coated AuNps for delivering the natural anticancer compound T-res and on characterization, a peak at 526.4 nm indicated the presence of bare gold nanoparticles, whereas SA coated nanoparticles showed a blue shift that is decrease in peak. This was because the SA improved the nanoparticles dispersion and stability and reduced the interparticle interactions or aggregation. Absorption intensity increases with heat due to improved SPR While analyzing AuNPs, a blue shift was observed for a smaller particle and greater pH resulting in increase in the peak, i.e., from 530 to 640 nm. Overall, UV–vis is an efficient method as it does not require any sample preparation and gold nanoparticles can be analyzed straight way after synthesizing.

7.

7

UV–vis absorption spectra of Au NPs (5 nm) (A), Au NPs (20 nm) (B), and sodium tetrachloroaurate and gold nanoparticles (C). Panel (A) and (B) Reproduced from ref . Copyright 2025, with permission of Elsevier; Panel (C) Reproduced from ref . Copyright 2023, with permission of MDPI.

2.4.2. Fourier Transform Infrared Spectroscopy (FTIR)

This characterization technique identifies the presence of functional group. In the case of gold nanoparticles synthesized from plant extracts, this method showed the functional groups involved in reduction, capping and stabilization of gold nanoparticles. It also confirmed the presence of phytochemicals by showing a common peak at 3389 cm–1 indicating O–H stretch, alcohol, 2919–2844 cm–1 C–H stretch, 1458 cm–1 (N–H bending), 1700 cm–1 (CC stretch). This is a highly sensitive method for nanoparticles. Nowadays FTIR also shows the interaction of gold nanoparticles with the reactive agents in few seconds. Similarly, advanced version is called attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR), which is extremely useful for nanoparticles as it helps in understanding the composition and behavior of these materials. Key absorption bands include: 3286 cm–1, attributed to −OH stretching vibrations of phenolic compounds and other phytochemicals; 2928 and 2875 cm–1, corresponding to −CH stretching of alkanes; 1636 cm–1, associated with −NH bending and −CO stretching; 1512 cm–1, related to −CH vibrations of alkanes and −NO stretching of nitro compounds; 1454 cm–1, indicative of −OH bending and – CO stretching in phenolics and similar compounds; and 1387 cm–1, assigned to −CN stretching of aromatic amine groups (Figure A).

8.

8

(A) ATR-FTIR spectrum and (B) XRD spectrum of the synthesized Au NPs. Reproduced from ref . Copyright 2022, with permission of MDPI.

2.4.3. Atomic Force Microscopy

Commonly known as AFM, is a technique used to determine the structural properties of gold nanoparticles via three-dimensional (3D) image showcasing particle’s size, shape and surface feature. This technique is vital for their use in different fields. When this technique was performed on untreated, gold-coated silanized glass plates with scans at 2.0 Hz with 256 pixels per line. AuNPs appeared roughly spherical with an initial average diameter of 100 nm with a height profile approximately 409 nm after extended soaking. Surface roughness for silanized glass without gold was observed to be 0.755 nm which increases with gold deposition showcasing the nanoparticle attachment. Maximum roughness is observed at 2 h. AFM can be performed in various environments including ambient, gas, liquid and requires minimum surface preparation. Its only disadvantage is its limited scanning size. However, it also provides insights about the ligand binding at nanoscale making it one of the most vital methods. Figure revealing individual spherical gold nanoparticles (AuNPs) and their aggregates. The measured average diameter of the AuNPs ranged from approximately 11 to 19 nm.

9.

9

AFM phase images of AuNPs on a bare graphite substrate. The circled regions exhibited gold nanoparticles and their diameter (particles around 11–19 nm). Reproduced from ref . Copyright 2023, with permission of MDPI.

2.4.4. X-ray Diffraction (XRD)

This technique is performed to identify the crystal structure, phase and spatial arrangement of an atom. The X-ray is irradiated on the gold nanoparticle, and the intensity of scattering angle of X-ray from gold nanoparticle is measured. For gold nanoparticles it shows a distinct peak which matches the cubic structure of gold (Figure B). If a broad peak appears then it indicates the smaller particle size because of the quantum confinement. It is an important characterization technique as it helps to find out the nanoparticle size and structural changes via laser energy. XRD for gold nanoparticle entrenched in a silica xerogel matrix showcased the crystalline nature of the gold nanoparticles, displaying peaks at 2θ = 38.21, 44.45, 65.04, and 77.65° which is corresponding to the (111), (200), (220), and (311) planes of face-centered cubic (FCC) gold. These peaks confirmed the formation of metallic and crystalline AuNps. But on XRD of AuNPs coated with SA, we could observe no significant change in the crystal structure. Further to estimate the particle size, Debye–Scherrer equation should be used. Danchova et.al, study showcased the XRD of AuNPs in a nanocomposite of silica, proving crystalline AuNPs at 111 reflections. It also showed weak peaks of Au-thiolate complexes and at high temperature low cristobalite phase was formed. Similar results were obtained when C. limon juice and E. prostrata leaves were used as reducing agents along gold nanoparticles and a constant, strong peak around 534 nm was observed in all such cases. Overall it is a very versatile technique and as per the XRD card (JCPDS no. 65–2870), the prominent peak at 20 = 38.31, 44.47, 64.58 and 77.43°, indexed to the planes of (111), (200), (220), and (311) is for gold nanoparticles. Only disadvantage of this technique is the XRD peaks are too broad for particles with size less than 3 nm and it cannot detect the amorphous materials.

2.4.5. Scanning Electron Microscopy (SEM)

Commonly called SEM is a technique used to identify the morphology and size of particles. It generates three types of images - external X-ray maps, backscattered electron images, and secondary electron images.

It needs simple sample preparation by putting a drop of solution on silica plane other conductive substrates and then removing the excess solution. In case of gold nanoparticles to confirm its presence, we can observe spherical particles and enlarged size in SEM images due to the laser energy. It can show variety of geometry like square, rectangle, cubic, triangular all nearly at 60 nm diameter.These nanoparticles can be easily analyzed as SEM does not need any intense sample preparation. It also shows the surface texture, structure and composition. When gold nanoparticles were synthesized using knotweed extract in Pulit et.al, study, we could clearly see spherical rod like structures ranging from 20–200 nm in SEM image indicating the presence of AuNPs. Similarly, gold nanoparticles synthesized using Nepenthes khasiana leaf extract, showed cluster with particle sizes between 50–80 nm. Overall, green synthesized gold nanoparticles tend to form spherical shapes, irrespective of any conditions. Only limitation is that it does not give any details on internal structure and is a very labor intensive and expensive technique. Figure displays the obtained images for tetrachloroaurate­(III) dihydrate (NaAuCl4) appears as cubic crystals. At a magnification of 30,000×, two crystals measuring approximately 0.38 and 0.67 μm were observed.

10.

10

SEM images at 2000× and 30,000× magnification of NaAuCl4 and AuNP samples. Reproduced from ref . Copyright 2023, with permission of MDPI.

2.4.6. X-ray Photoelectron Spectroscopy (XPS)

This spectroscopy is usually used to analyze the surface composition and chemical state of gold nanoparticles (AuNPs) immobilized on silanized glass substrates. Sharp peaks for Au 4f, Au 4d, and Au 4p and Au 4f7/2 at 84 eV, confirms the presence of gold. It has a superior capacity to ascertain chemical states, extensive applicability, and nondestructive characteristics. XPS applications encompasses the assessment of oxidation states in nanoscale particles. It also offers insights on nanoparticle surfaces and chemistry, majorly in comparison to uncoordinated ligands. In Seit’s study, there was a significant increase in oxygen-containing carbon species on gold-coated samples, which proves the chemical modification due to the presence and attachment of AuNP. In Tendo et.al, study XPS confirmed the chemical state and single layer orientation of flat gold films and AuNPs. It also helps to identify the effective molecular layer thickness by comparing C 1s and Au 4f peak intensities and gives 10 Å (MP) and 19 Å (MBP) confirming the AuNP films. Similarly on narrow scans for AuNPs synthesized via bacteria, peaks were visible with binding energy between 84 to 88 eV corresponding to aryl shell features. Furthermore, for AuNPs made from Ulva linza, Au was clearly observed due to the presence Au 4f7/2 and Au 4f5/2 peaks. Carbon, Nitrogen, Sulfur was also detected, which came from the Ulva linza and helped in reduction of gold ions to nanoparticles to prevent surface clumping and interaction. In some studies, where gold nanoparticle was capped with cysteine or glutathione (that is thiol containing compounds) XPS showed S 2p binding energies (161–164 eV), confirming strong Au–S bonding, essential for nanoparticle stabilization. It also detects the surface contamination by detecting elements like Cl, Na, or C. Furthermore, in bimetallic nanoparticles like Au–Ag or Au–Pd, XPS differentiates interaction and elemental distribution by showcasing shell composition or surface enrichment of one metal over the other. Various studies indicate that, AuNPs synthesized in a green manner showcases peaks with higher binding energy due to surface oxidation and interaction. The prominent shift in peaks denotes the partial oxidation or rigid, stubborn, stable capping interaction.

2.4.7. Transmission Electron Microscopy (TEM)

TEM is widely used to study the internal structure, surface morphology, shape, particle size distribution and dispersion of gold nanoparticles. Since electrons have much shorter wavelengths than light, TEM is ideal as it gives better resolution for nanoparticles with nano sizes (Figure ). Its advanced version is the High-resolution TEM (HRTEM) which shows clear images of the gold core and helps to identify whether particles are crystalline or amorphous. To operate, this method requires a sample preparation, which may lead to gold NP aggregation. Surface alterations are undetectable by normal or ordinary TEM techniques and to overcome this issue glycerol spraying/low-angle rotating metal shadowing TEM and cryo-TEM, is necessary. For AuNPs, a 5–100 nm, spherical shapes are observed on TEM images. Usually polydispersity is confirmed at 30–100 nm range and on heating, gold nanoparticles agglomerate making it easy to identify on TEM images. Similarly, TEM analysis of AuNPs synthesized via bacteria, showed spherical and triangular sites and these nanoparticles synthesized at different conditions had different sizes. For example, AuNPs synthesized at 25 °C had a size of approximately 39.0 ± 9.1 nm and for 42 °C, 36.7 ± 7.7 nm. It was noticed that at pH 12.7, the size was decreased. Recently, AuNPs were synthesized from T. farfara flower buds and was charactersied using TEM which revealed particle size 15–20 nm, indicating efficient size control via plant extract. Overall, TEM images prove that the green-synthesized GNPs are usually spherical, but also show hexagonal, pentagonal, and triangular shapes.

11.

11

TEM images of Au NPs (5 nm) (A, B) and Au NPs (20 nm) (C, D). Reproduced from ref . Copyright 2025, with permission of Elsevier.

2.4.8. Dynamic Light Scattering (DLS)

Dynamic Light Scattering is one of the common and widely used technique to identify the size distribution, average particle size and polydispersity index (PDI) of gold nanoparticles dispersed in a colloidal medium. Unlike electron microscopy techniques that provide direct imaging, DLS offers hydrodynamic diameter measurements that include the particle core, surface coating like capping agents and solvent layer. This makes it valuable for tracking size changes during synthesis, bioconjugation, or storage. This method is based on simple principle which analyses the intensity of light scattering when nanoparticles undergo Brownian motion in suspension. Another study reported synthesis of AuNPs using Evolvulus alsinoides extract and on characterization, the DLS revealed sizes ranging from 50–100 nm with a dominant size near 80.29 nm. Similarly, Mapala et al. found that Mimosa pudica flower extract-refereed AuNPs showed an average size of 24 nm with a narrow range (15–62 nm), which was later confirmed by TEM analysis. DLS also monitors the gold nanoparticles stability by measuring the ζ-potential, which reflects surface charge. High absolute ζ-potential values (typically more than or equal to ± 30 mV) indicate strong interparticle repulsion and colloidal stability, while low values suggest potential aggregation making it advantageous. DLS analysis of AuNPs reported by Oliveira et al. is depicted in Figure . But it may overestimate size in polydisperse or aggregated samples and assume spherical shape during analysis, making other techniques like TEM or SEM essential for accurate morphological validation.

12.

12

(a) DLS analysis of particle size distribution and ζ-potential and (b) schematic representation of the double layer that surrounds the nanoparticle in aqueous medium. Reproduced from ref . Copyright 2023, with permission of MDPI.

2.4.9. Energy-Dispersive X-ray Spectroscopy (EDX)

This technique is widely used to detect the elemental composition of gold nanoparticles (AuNPs). It provides qualitative, semiquantitative, and quantitative data by detecting X-rays emitted from the sample during electron beam interaction and is often performed with electron microscopy like SEM or TEM. EDX also helps in plotting the spatial distribution of other elements present, making it easy to identify the presence of gold. It is major disadvantage is precision of chemical analysis and quantification which is often used in conjunction with X-ray diffraction (XRD) for a more complete understanding of nanoparticle composition and crystallinity. Hence, a combination of XRD and EDS is the best for characterization of gold nanoparticle. For gold nanoparticles and ions, EDX shows a signature peak near 2 to 3 keV. Some reports also show an additional peak near 9 keV, depending on the instrumentation and settings. For AuNPs mediated using T. argentiea flower extract, the EDX depicted a strong gold peak at 3 keV, along with minor signals from carbon, oxygen, and the gold content was estimated to be around 52.27%. ,,

3. Applications of AuNPs

3.1. Antimicrobial Activity of AuNPs

A number of applications of AuNPs have been shown in Figure . The growing resistance of pathogenic microorganisms to a wide range of antibiotics, including the latest ones, presents a major challenge in clinical medicine. One potential solution lies in the use of metal nanoparticles, particularly gold nanoparticles (AuNPs). Gold Nanoparticles are widely used for antimicrobial applications (Table ) due to their small sizes, high surface area, and unique ability to interact with microbial cells. The antimicrobial effectiveness (Figure ) of AuNPs is influenced by factors such as the synthesis method, particle size, shape, and the concentration of biologically synthesized nanoparticles. One of the crucial and unique properties of AuNPs is its positively charged nature, which enables them to interact with Gram-positive and Gram-negative bacteria. However, due to their thinner cell walls, Gram-negative bacteria are more susceptible to AuNP penetration. In contrast, Gram-positive bacteria possess a thick peptidoglycan layer that acts as a barrier, limiting nanoparticle entry. , Gold nanoparticles supported on clinoptilolite, mordenite, and faujasite zeolites effectively eliminated E. coli and Salmonella typhi. Faujasite-supported AuNPs (5 nm) showed the highest dispersion and efficiency, reducing bacterial colonies by 90–95%. The zeolite support significantly influenced nanoparticle size, roughness, and biocidal activity. Functionalized gold nanoparticles (AuNPs) effectively combat multidrug-resistant (MDR) bacteria, targeting both Gram-negative and Gram-positive uropathogens. Cationic and hydrophobic AuNPs suppressed 11 MDR clinical isolates with minimal toxicity to mammalian cells. Their surface chemistry plays a crucial role in antimicrobial activity, offering a promising long-term strategy against bacterial resistance.

13.

13

Multifield applications of AuNPs.

8. Antimicrobial Activity of AuNPs.

Sr. No. gold nanoparticle type microbe species efficacy/activity application references
1 Gold Nanostars (AuNSTs) E. coli, S. aureus ZOI: 20.0 ± 0.54 mm (E. coli), 23.0 ± 0.35 mm (S. aureus); MIC: 1.25 μg/mL (E. coli), 0.625 μg/mL (S. aureus) Antibacterial treatment
2 Actinomycetes-based Gold Nanoparticles (Ac-AuNPs) Streptomyces sp. ASM19 IC50: 3.77 mg/mL (SCC9), 1.56 mg/mL (SCC25); Induced apoptosis (26.37% SCC9, 32.08% SCC25); G2/M cell cycle arrest Biomedical applications (nano drug delivery, anticancer therapy)
3 Cassia sericea-derived AuNPs S. aureus, Pseudomonas aeruginosa, E. coli 82.45% wound closure (antibacterial and wound healing) Antimicrobial & wound healing agent
4 AuNRs@PDA@AgNPs nanocomposites E. coli, S. aureus MIC: 16 μg/mL; 5.3 and 2.0% bacterial viability for E. coli & S. aureus Synergistic photothermal and chemical antimicrobial agent
5 AMP-AuNCs (Antimicrobial Peptide-functionalized Gold Nanoclusters) E. coli, six common bacterial species 100% accuracy in bacterial ID, 86.7% accuracy for clinical E. coli isolates Rapid bacterial diagnostics for clinical applications
6 AuNP/WO3 NPL/r-GO ternary heterostructure Carbapenem-resistant E. coli (CRE E. coli), Methicillin-resistant S. aureus (MRSA) 100% inactivation of CRE E. coli and MRSA superbugs after 60 min of sunlight exposure Photothermal-photocatalytic antimicrobial and antibiotic degradation agent
7 Polyphenol Nanoparticles (PNPs) Micrococcus luteus, E. coli Growth inhibition: 50.7% (M. luteus), 12.1% (E. coli) Antimicrobial coatings to prevent biofilm formation
8 HBPAA-capped Ag@AuNPs-coated cotton fabric E. coli, S. aureus MIC: 5 mg/L; 100% bactericidal efficacy even after 50 washes with 900 mg/kg Ag@Au content Washable antimicrobial textiles with organic–inorganic coupling antibacterial properties
9 Cyphostemma adenocaule (CA)-AgNPs S. aureus, E. coli 88% S. aureus biofilm inhibition (125 μg/mL), broad-spectrum antibacterial activity Antibacterial, antibiofilm, antioxidant agent for food, cosmetics, and medicine
10 Iris kashmiriana-derived AuNPs E. coli, S. aureus, Pseudomonas aeruginosa, Streptococcus mutans MIC-based antibacterial activity, ROS-mediated mechanism, antibiofilm and antiadherence effects Antibacterial coatings for orthodontic appliances (wires and brackets)
11 Eisenia bicyclis-gold nanoparticles (EB-AuNPs) Listeria monocytogenes, S. aureus, Pseudomonas aeruginosa, Klebsiella pneumoniae, Candida albicans MIC: 512–2048 μg/mL; Biofilm inhibition: 91.13% (S. aureus), 58.60% (K. pneumoniae), 57.22% (P. aeruginosa), 33.80% (L. monocytogenes) Antibacterial, antibiofilm, antivirulence agent against bacterial and fungal pathogens

14.

14

Mechanism of antibacterial action of AuNPs and unreduced gold ions. Reprinted with permission from ref . Copyright 2022 MDPI.

3.2. Antioxidant Activity of Gold Nanoparticles

A study by Katas et al. showed powerful antioxidant effects toward AuNPs, helping to protect cells from harmful free radicals. This ability can reduce the oxidative stress-related damage. It contributes to aging and diseases like cardiovascular and neurodegenerative treatments. Gold nanoparticles (AuNPs) stabilized with carboxymethylated frankincense resin (CMFR) showed antioxidant activity by neutralizing DPPH free radicals. AuNPs synthesized from CMFR-AuNPs (7–10 nm) displayed dose-dependent antioxidant properties. Due to the large surface area and strong stability free radical scavenging. CMFR-AuNPs showed potential therapeutic advancements. For instance, a study by Faraday et al. examined that gold nanoparticles affect light transmission and color based on their size and dispersion. The study also examined the gold’s stability and reactivity in different conditions, which shows antioxidant behavior. Gold nanoparticles (AuNPs) produced through the electrodeposition were evaluated using the DPPH assay. Suliasih et al. found that smaller AuNPs with greater particle density exhibited higher antioxidant activity. AuNPs synthesized at −1 V depicted the highest inhibition (36.80%) of free radicals. In another study, Ipek et al. synthesized Gold nanoparticles (AuNPs) using Allium cepa L. peel extract. The analysis identified their spherical shape, ranging from 6.08 to 54.20 nm. Antioxidant tests (DPPH, ABTS, and CUPRAC) showed moderate free radical scavenging ability, less effective than BHA and α-tocopherol. AuNPS synthesized from Spirulina platensis depicted strong antioxidant activity. The DPPH assay showed that at 300 μg/mL, achieved 70% inhibition, effectively neutralizing free radicals. The results showed Spirulina-based AuNps as a natural antioxidant with potential benefits in biomedical and pharmaceutical fields. Another study compared the antioxidant properties of chitosan and green-synthesized gold nanoparticles (AuNPs). Chitosan acted as both a reducing and stabilizing agent for AuNPs and was measured using DPPH, ABTS, hydroxyl radical scavenging, and FRAP assays. Low molecular chitosan (47.8 kDa) showed the highest antioxidant effect. Another study by Radadi et al. showed AuNPs synthesized from Annona muricata leaf extract through ABTS assay, with a maximum of 96.4 μg/mL. Characterization of nanoparticles using UV–vis, FTIR, TEM, and XRD exhibited spherical shape and crystalline structure. Their antioxidant properties are linked to polyphenols and flavonoids in the extract, which enhance the free radical scavenging. Gold nanoparticles (AuNPs) synthesized using Vibrio alginolyticus were tested using DPPH and metal chelating assays. Characterization depicted irregular shapes, ranging from 100 to 150 nm. The nanoparticles showed greater antioxidant effectiveness at lower concentrations. It emphasizes the eco-friendly synthesis of AuNPs. For instance, another study of AuNPs synthesized using Alternaria chlamydospore was tested using the DPPH assay, achieving 71.2% inhibition at 500 μL. Nanoparticles confirmed their spherical shape ranging from 12–15 nm with a high surface area-to-volume ratio, enhancing free radical neutralization. The IC50 value was 224.97 μg/mL, demonstrating strong antioxidant activity. Gold nanoparticles (AuNPs) results suggest that they could be valuable for biomedical applications.

3.3. Anti-Inflammatory Activity

Gold NPs have been extensively used for their anti-inflammatory benefits. Synthesis is done by using various methods such as green synthesis with plant extracts, microbial sources, and chemical modifications. For instance, Au-NPs synthesized using Saussurea costus extract exhibited strong anti-inflammatory effects due to the presence of bioactive polyphenols that stabilize and enhance their biological activity. Similarly, Capsicum annum fruit extract-mediated Au-NPs showed notable anti-inflammatory and antiangiogenic properties. Au- NPs derived from Chaetomium globosum extract demonstrated potent inhibition of inflammatory mediators, making them a viable option for anti-inflammatory treatment. Sivakavinesan et al. synthesized Au-NPs via Citrus sinensis fruit peel extract, which exhibits strong antioxidant and anti-inflammatory activities Similar study was conducted by Gao et al. in which gold nanoparticles were synthesized using citrus peel extract with the ultrasound-assisted method and showed enhanced anti-inflammatory activity. Additionally, AuNPs synthesized from papaya peel extract demonstrated antimicrobial, antioxidant, and anti-inflammatory effects, reinforcing their relevance in pharmaceutical and biomedical fields In a study by Elizalde-Mata et al. AuNPs derived from Croton draco extract exhibited strong antioxidant and anti-inflammatory properties, making them promising for therapeutic applications. Similarly, Eltahir et al. reported that phenolic compounds from Glycyrrhiza glabra showed notable anti-inflammatory effects, suggesting their potential in treating inflammation-related disorders. In another study conducted by Nasab et al. conjugated Au-NPs with cortistatin peptides were successfully employed for targeted drug delivery to asthmatic lung tissues, offering a novel approach for respiratory inflammatory diseases. The Saussurea Costus gold nanoextract demonstrated significant antioxidant, antidiabetic, anti-Alzheimer, and anti-inflammatory activities, making it a potential therapeutic agent for managing oxidative stress related diseases. Prasad et al. synthesized gold nanoparticles using an aqueous extract of Commiphora wightii which were potential anti-inflammatory agents. Hongsa et al. developed chitosan-collagen coated gold nanoparticles for targeted drug delivery of 5-fluorouracil (5-FU), a chemotherapy drug that exhibited controlled and sustained drug release, anti-inflammatory activity, and cytotoxicity studies confirmed their anticancer potential

3.4. For Skin Disease Treatment

Gold nanoparticles (AuNPs) have emerged as a promising tool in skin disease treatment due to their biocompatibility, enhanced skin penetration, and controlled drug release properties. They offer therapeutic benefits for conditions like psoriasis, atopic dermatitis, skin cancer, and microbial infections by reducing inflammation, stabilizing the skin barrier, and enhancing drug delivery. Qui et al. successfully synthesized antibacterial photodynamic gold nanoparticles (AP-AuNPs) for treating skin infections. These nanoparticles, combining antibacterial peptides and photodynamic effects, showed significant antibacterial activity against S. aureus and E. coli, while also promoting wound healing in infected skin tissues. Cuyler et al. explored the potential of gold nanoparticles synthesized using Bulbine Frutescens to treat Eczema. The nanoparticles demonstrated significant wound closure and inhibited histamine production, suggesting their potential to alleviate skin damage associated with atopic dermatitis Gold nanostructures can also be implemented in melanoma therapy. Leu et al. studied the effect of AuNPs combined with antioxidant epigallocatechin gallate and α-lipoic acid on wound healing in mice, demonstrating anti-inflammatory and antioxidative effects Another study conducted by Oliveira et al. utilized AuNPs in combination with photobiomodulation (low-power laser therapy) to treat dermonecrotic lesions, specifically those caused by the venom of the spider Loxosceles simillis. It effectively reduced necrotic tissue and erythema in animal models Poomrattanangoon and Pissuwan evaluated the wound healing ability of gold nanoparticles coated with collagen-I. Collagen-I@AuNPs reduced the levels of inflammatory cytokines and induced the growth factors involved in wound healing. Dong et al. developed mussel-inspired electroactive, antibacterial, and antioxidative composite membranes with gold nanoparticles and antibacterial peptides for enhancing skin wound healing. The composite membranes promoted cell proliferation and migration and exhibited enhanced antibacterial and antioxidant effects Wu et al. developed an injectable, antibacterial hydrogel based on gold nanorods and an N-calamine polymer for bacteria-infected skin wound healing. The hydrogel demonstrated excellent in vitro antibacterial capacity and promoted wound healing Abbas et al. investigated the efficacy of alginate-coated gold nanoparticles against antibiotic-resistant Staphylococcus and Streptococcus strains that cause acne. The results concluded the potential of Gold NPs as a potential antimicrobial agent to combat antibiotic resistance in acne treatment

3.5. Ocular Disease Treatment

Gold has long symbolized nobility and attracted attention due to its shiny appearance, it has excellent ductility, biocompatibility, and molecular recognition, and gold is widely used in various fields. Gold nanoparticles (AuNPs) exhibit unique physical and chemical properties, enabling biomedical applications. AuNPs easily bind to proteins and antibodies, aiding in disease diagnosis, gene detection, and cancer treatment. They also show cytotoxic effects on cancer cells and possess antioxidant and anti-inflammatory properties. Research highlights their therapeutic potential in treating retinopathy, neurological diseases, cancers, cardiovascular diseases, infections, and metabolic disorders. Delivering drugs through the ocular route holds great potential for treating various eye diseases, including diabetic retinopathy. This condition is primarily driven by inflammation and elevated vascular endothelial growth factor (VEGF) levels, leading to abnormal blood vessel growth (neovascularization). Ocular drug delivery systems aim to effectively target these factors, improving treatment outcomes for such vision-threatening disorders. Ocular drug delivery is an effective method for treating eye diseases like ocular neovascularization in diabetic retinopathy, which is caused by inflammation, retinal ischemia, and the accumulation of advanced glycation end-products. Elevated vascular endothelial growth factor (VEGF), interleukins, and reactive oxygen species (ROS) also contribute to disease progression. Gold nanoparticles (GNPs) possess antioxidant and antiangiogenic properties, making them ideal for ocular drug delivery. They are biocompatible, easy to synthesize, and can be functionalized to improve movement across ocular barriers. Apaolaza et al. conducted a study where low molecular weight hyaluronan (HA) was used to enhance nanoparticle stability, mobility, and targeting via CD44 receptor interaction. HA-GNPs effectively reached deeper retinal layers, inhibiting neovascularization and protecting retinal pigment epithelial cells. Despite a slight reduction in antioxidant activity, longer studies are needed to assess their full potential in delivering antiangiogenic treatments for intraocular vascular diseases. HA-AuNPs showed excellent colloidal stability, with the HA coating preventing aggregation in the trabecular meshwork of ex vivo porcine eyes. Nanoparticles (120 nm) accumulated the most in this region. Advancements in nanotechnology have improved nanoparticle stability, while coatings like poly­(ethylene glycol) (PEG) enhance biocompatibility, reducing toxicity and increasing circulation time. Although AuNPs show significant potential in biomedical applications, further safety studies are required before clinical implementation. Further research is needed to clarify ocular distribution and potential toxicity, as current biocompatibility data remain conflicting.

3.6. For Diabetes Treatment

Diabetes mellitus, commonly known as diabetes, occurs due to reduced insulin secretion by the pancreatic islet cells, leading to elevated blood glucose levels (hyperglycemia). It is characterized by symptoms such as unexplained weight loss, excessive urination (polyuria), increased thirst (polydipsia), and an excessive. Diabetes mellitus is classified into Type 1, Type 2, and Gestational diabetes. Type 1 diabetes results from an autoimmune attack on pancreatic β cells, leading to insulin deficiency. For children, the term juvenile diabetes is used. Type 2 diabetes is caused by insulin resistance and reduced insulin secretion due to defective insulin receptor response. Gestational diabetes occurs during pregnancy when hormonal changes reduce insulin sensitivity and increase blood sugar levels. Management includes dietary modifications, exercise, insulin therapy, and oral medications. Recent nanomedicine research focuses on using nanoparticles for effective insulin delivery in Type 1 diabetes treatment. Nanoparticles offer promising advancements in controlled insulin release, improved bioavailability, and fewer side effects, providing a potential breakthrough in diabetes management and treatment strategies for better patient outcomes. Gold nanoparticles (AuNPs) have shown potential as therapeutic agents for diabetes treatment. As they exhibited anti-inflammatory, antioxidant, and antihyperglycemic effects, disrupting key disease pathways. However, their safety, optimal size, and dosage require further detailed study. Proper characterization is essential to ensure safe applications for treating diabetes and related microvascular complications. Daisy et al. prepared an aqueous extract using Cassia fistula stem bark and synthesized gold nanoparticles (AuNPs) to assess their hypoglycaemic effects. Synthesized AuNPs were characterized using spectroscopy and electron microscopy. In streptozotocin-induced diabetic rats, AuNPs significantly lowered blood glucose, improved liver and kidney function, enhanced lipid profiles, and increased body weight more effectively than the aqueous extract. Phytochemically synthesized AuNPs demonstrated excellent antidiabetic properties by stabilizing serum biochemistry and reversing renal dysfunction. This study confirms that Cassia fistula-derived AuNPs are promising therapeutic agents for diabetes mellitus treatment, outperforming conventional plant extracts in improving metabolic health. AuNPs also reduces hyperglycaemia, oxidative stress, inflammation, and the proteolytic pathway. Gold nanoparticles (AuNPs) in conjugation with natural products have demonstrated promising antidiabetic properties, such as reduction in glycated hemoglobin levels and anti-inflammatory effects. Opris et al. conducted a study aimed to evaluate the therapeutic potential of AuNPs functionalized with Sambucus nigra L. (SN) extract in an experimental rat model of diabetes. Diabetes was induced in 18 male Wistar rats using a single streptozotocin injection. The diabetic rats were then treated with SN extract, gold nanoparticles (AuNPs), or saline for 2 weeks via oral gavage. Another 18 nondiabetic rats received the same treatments. After treatment, blood, liver, and muscle samples were analyzed for oxidative stress markers, liver MMP-2/-9 activity, COX-2 and NFKB expression, and histopathological changes. Serum glucose, cholesterol, ALAT, and ASAT levels were also measured to assess the effects of AuNPs and Sambucus nigra L. extract on diabetes-related metabolic and inflammatory changes. The administration of AuNP-SN extract significantly increased the muscle and systemic GSH/GSSG ratio in diabetic rats compared to untreated diabetic (p < 0.03) and nondiabetic vehicle-treated groups (p < 0.05). Malondialdehyde (MDA) levels were reduced in the AuNP-treated diabetic group compared to nondiabetic controls (p < 0.05). Additionally, COX-2 expression (p < 0.0001) and proMMP-2 activity (p < 0.05) were decreased, along with a significant reduction in Kupffer cell percentage (<0.001). Histopathological analysis revealed no structural abnormalities in liver tissue. These findings suggest that AuNPs functionalized with Sambucus nigra L. extract possess strong potential as adjuvants in diabetes therapy by enhancing antioxidant defenses, reducing MMP activity, and mitigating inflammation in liver tissue. Further research is warranted to explore their clinical applications in diabetes management.

3.7. Cardiovascular Disease Treatment

Gold nanoparticles (AuNPs) are now being widely used for treating cardiovascular diseases (CVD) due to their ability to increase the drug delivery to a targeted site, reducing side effects, and maximizing bioavailability (Table ). AuNPs which are in functionalized form are capable of delivering drug molecules to targeted tissues with enhanced specificity, offering controlled release as well as longer action. These nanoparticles are ideal for controlling atherosclerosis, thrombosis, and myocardial infarction and provide a probable pathway for prospective cardiovascular therapies due to their anticoagulant, antioxidant, and anti-inflammatory properties. Second, due to their unique optical, electronic, and biological properties, they also show potential in recovering cardiovascular disease. They can target selectively atherosclerotic plaques, provide controlled drug release, and function as imaging agents for early diagnosis. They can also decrease oxidative stress, regulate inflammatory reactions, and enhance endothelial function, thereby providing cardio protection. Among the diverse theragnostic nanomaterials, gold nanoparticles (AuNPs) are remarkable because of their special optical and physicochemical properties. AuNPs can facilitate the diagnosis of CVDs by computed tomography (CT) and can carry out photothermal therapy to induce plaque ablation. Their therapeutic efficacy with a reduced side effect is increased due to the ability to deliver drugs directly to the infected effects. Nanotechnology here plays a critical role in the improvement of CVD management as these particles increase therapeutic efficiency, minimize side effects, and allow for controlled drug release. On using branched polyethylenimine-coated gold nanoparticles (bPEI-AuNPs) in collagen hydrogels, it greatly improves the drug delivery, mechanical properties, and conductivity. These hydrogels also improve cardiomyocyte function, inducing rhythmic and synchronized beating. In fact, on incorporating gold nanoparticles (AuNPs) into the biosensor architecture, sensitivity and specificity increase due to their conductivity, extensive surface area, and biocompatibility. The performance of the biosensor, such as its low detection limit (LDL) and high specificity, shows its promise for clinical use, offering a reliable means for evaluating cardiovascular risk and individualized treatment plans. The measurements indicated remarkable blood velocity, pressure, and temperature variations, and additionally, the in vivo biodistribution of AuNP-miR-67, administered subcutaneously, indicated the particles were being cleared mainly through the liver and kidneys within 11 days. Ven et. al points to the possibility of the PNP-AuNP-miRNA hydrogel platform in controlled, sustained miRNA delivery with the additional advantage of being minimally invasive. Future advancements can include the inclusion of targeting ligands for cell-specific delivery, and increasing therapeutic efficiency for targeted treatment of cardiovascular disease and other conditions.

9. Role of AuNPs in Cardiovascular Disease Treatment.

cardiovascular disease role of AuNPs mechanism of action references
Atherosclerosis Targeted Drug Delivery, Plaque Reduction, Anti-inflammatory Effects Functionalized with anti-inflammatory agents and statins to reduce cholesterol, inhibit foam cell formation, and stabilize plaques.
Myocardial Infarction (MI) Cardiac Tissue Regeneration, Drug Delivery, Theranostics Loaded with growth factors, cytokines, and drugs to enhance tissue repair, reduce apoptosis, and promote angiogenesis.
Hypertension Antihypertensive Drug Delivery, Vasodilation Functionalized with NO donors for sustained vasodilation and endothelial function improvement.
Thrombosis Thrombolytic Therapy, Clot Dissolution Functionalized with thrombolytic agents for targeted clot lysis and prevention of rethrombosis.
Ischemia or Reperfusion Injury Reduction of Oxidative Stress Conjugated with antioxidants and antiapoptotic agents to scavenge ROS, reduce inflammation, and prevent cell death.
Coronary Artery Disease Diagnostic Imaging, Plaque Detection, and Treatment AuNPs enhance imaging via CT, MRI, and photoacoustic techniques for precise localization and treatment.
Arrhythmias Electrophysiological Regulation, Drug Delivery Conjugated with antiarrhythmic drugs to regulate cardiac rhythm and prevent arrhythmias.
Stroke Thrombolytic Therapy, Neuroprotection AuNPs conjugated with neuroprotective agents to reduce brain damage and restore blood flow.
Venous Thromboembolism Clot Targeting, Thrombolytic Therapy AuNPs functionalized with anticoagulants to prevent clot formation and enhance clot dissolution.
Cardiovascular Imaging Enhanced CT and MRI Imaging, Real-time Monitoring AuNPs provide high-contrast imaging for cardiovascular diagnosis, identifying plaques and occlusions.
Pulmonary Hypertension Antihypertensive Drug Delivery, Vasodilation Functionalized with vasodilators and NO donors for targeted therapy.

3.8. Anticancer Applications

Gold nanoparticles (AuNPs) have become attractive nanoplatforms for cancer diagnosis and therapy because of their size, shape, and optical tunability (Table ). Their use in photothermal therapy, drug delivery, and imaging has exhibited increased tumor targeting (Figure ) with low systemic toxicity. Yet, issues like biodistribution, cytotoxicity, and immunogenicity must be explored before they can be translated to the clinic. Optimizing AuNP formulations to enhance efficacy and safety in cancer treatment will be a task for future studies. These nanoparticles play a vital role as an anticancer due to their size, shape and physiochemical properties they inhibit. Properties like biocompatibility, high surface area to volume ratio and ease on functionalization makes them suitable for delivery of chemotherapeutic agents on tumor sites. They also help in minimizing the damage to healthy cells and reducing systemic toxicity. By offering controlled drug release, targeted delivery, and reduced side effects, gold nanoparticles represent a multifaceted tool in advancing more effective and personalized cancer treatments. Gold nanoparticles are ideal carriers for targeted drug delivery as they passively accumulate in tumor tissues due to leaky vasculature and poor lymphatic drainage. This is due to an effect known as ‘Enhanced Permeability and Retention Effect’. Second, due to their strong optical properties, they are widely used in CT, MRI, and photoacoustic imaging which are all real-time imaging techniques. Some of its applications in different types of cancer are given in the table below.

10. Anticancer Potential of AuNPs.

type of AuNPs targeted cancer type observed effects references
Gold Nanorods modified with Silica Hepatocellular Carcinoma Induced localized hyperthermia leading to necrotic tumor cell death while sparing healthy tissue. ,
PEGylated AuNPs Loaded with miRNA Breast Cancer Restored tumor-suppressive miRNA expression, leading to reduced proliferation
Hollow Gold Nanospheres Brain Tumors Facilitated blood-brain barrier penetration and enhanced accumulation at tumor sites.
Resveratrol-loaded gold nanoparticles Pancreatic Cancer Induced apoptosis via mitochondrial pathway and reduced tumor volume in vivo.
AuNPs Functionalized with Anti-EGFR Antibodies Non-Small Cell Lung Cancer Enabled precision delivery and EGFR signaling inhibition, resulting in tumor regression.
Gold nanostructures Breast Cancer Synergistic photothermal and chemotherapeutic effects led to enhanced cell death.
Gold Nanoparticles coated on magnetic Fe3O4 nanoparticles Leukemia ultrasensitive and selective electrochemical detection of leukemia cells
Albumin conjugated Gold Nanoparticles Prostate Cancer moderately reduced adhesion in prostate cell lines suggesting potential to combat cancer.
Gold Nanoparticles Embedded in Liposomal Formulations Brain cancer enhanced brain delivery of docetaxel, achieving up to 4.08-fold higher accumulation than the marketed formulation Docel.
ScFv-Decorated Bimodal Nanoprobes from Au Breast Cancer Specific recognition and internalization in HER2-overexpressing cells; inhibited proliferation. ,
AuNPs (2–6 nm, microwave-synthesized) Breast (MCF-7), Colon (HCT-116) ROS-mediated cytotoxicity, impaired cell migration, upregulation of apoptotic genes, DNA damage
AuNPs synthesized using Antigonon leptopus extract Breast adenocarcinoma (MCF-7) GI50 = 257.8 μg/mL; inhibited cell growth; higher free radical scavenging activity than extract alone
Biogenic AuNPs synthesized using Brazilian Red Propolis (BRP) Bladder (T24), Prostate (PC-3) Dose-dependent cytotoxicity; highest effect from AuNP dichloromethane and AuNP extract; apoptosis-related cell death
Solanum xanthocarpum (Sx)-derived AuNPs Nasopharyngeal cancer Induced apoptosis via autophagy and mitochondrial pathway; reduced viability and colony formation
Shape-dependent AuNPs (rods, stars, spheres) Osteosarcoma; Pancreatic AuNPs stars showed highest cytotoxicity and anticancer potential; upregulated Bax, downregulated Bcl-2
Biosynthesized AuNPs (Curcumin, Turmeric, Quercetin, Paclitaxel conjugated) Breast cancer Synergistic inhibition of growth and angiogenesis
Mango seed–derived AuNPs General (in vitro) Moderate cytotoxicity and antioxidant activity
Sargassum glaucescens–stabilized AuNPs Cervical, Liver, Breast, Leukemia Induced intrinsic apoptosis (via caspase-3/9), no toxicity to normal cells
Mushroom-extract biosynthesized AuNPs Breast cancer Hexagonal AuNPs (∼13 nm) showed significant cytotoxicity
Combretum glutinosum based AuNPs Lung cancer Multishaped AuNPs showed cytotoxicity; also, part of a bi- and trimetallic system with catalytic and antibacterial properties
Nothapodytes fetida leaf extract AuNPs Osteosarcoma, Lung Cancer Spherical AuNPs showed significant anticancer and wound-healing properties
Curcumin-INH functionalized AuNPs (AuNPsCur@INH) Lung squamous carcinoma Showed selective cytotoxicity via ROS-mediated apoptosis
Panax notoginseng-mediated AuNPs Pancreatic cancer AuNPs induced ROS generation and intrinsic apoptosis
Pomegranate seed oil (PSO)-capped AuNPs Lung and colon cancer AuNPs (70 nm, +34 mV) in functional yoghurt reduced cell viability to ∼25–28%
Satureja rechingeri-mediated AuNPs Cisplatin-resistant ovarian cancer Spherical AuNPs (∼15.1 nm) induced apoptosis, cell cycle arrest (G0/G1), and gene regulation; effective against drug-resistant cells
Moringa oleifera leaf-extract AuNPs Breast cancer Exhibited anticancer activity with IC50 = 67.92 μg/mL
Ziziphus nummularia leaf-extract AuNPs Cervical and breast cancer Spherical AuNPs showed dose-dependent cytotoxicity to cancer cells, nontoxic to normal fibroblasts
Peptide-functionalized AuNPs (TAP@AuNPs) Breast cancer Targeted nucleolin receptors; induced apoptosis via cytochrome c and caspase-3/7 activation
Chemicaly synthesized AuNPs Bladder cancer upregulated Bax, downregulated Bcl-2 & VEGFA, reduced migration
Alternanthera sessilis-extract AuNPs Cervical cancer Induced cytotoxicity and apoptosis via intrinsic apoptotic pathway
Coleus scutellarioides leaf-extract AuNPs Breast cancer nontoxic to normal Hs-27 cells; also exhibited antioxidant activity
AuNPs loaded with copper(I) complexes General anticancer AuNPs improved bioavailability and controlled release of Cu(I)-based drugs; AuNPs–A system showed 90% drug loading and slow release over 4 days
Theaflavin-conjugated AuNPs Ovarian cancer Enhanced apoptosis via ROS generation and mitochondrial depolarization
Pm-AuNPs (from Pterocarpus marsupium bark) Oral squamous cell carcinoma Showed cytotoxicity in OSCC cell lines (IC50: 25–75 μg/mL); induced ROS generation, mitochondrial dysfunction
Dracocephalum kotschyi leaf AuNPs (d-GNPs) Leukemia (K562), Cervical (HeLa) Spherical AuNPs (∼11 nm) showed dose-dependent cytotoxicity (IC50: 196.32 and 152.16 μg/mL)
Trachyspermum ammi seed-extract AuNPs Liver cancer Spherical AuNPs induced ROS-mediated apoptosis
Magnolia officinalis-derived AuNPs Lung cancer Induced cytotoxicity and apoptosis by modulating apoptotic gene expression
Amygdalus communis (AC) leaf-extract AuNPs Colorectal (CaCo-2), glioma (U118), ovarian inhibited CaCo-2 proliferation
Crassocephalum rubens (AECR)-intermediated AuNPs Breast cancer; Colon Cancer showed significant dose- and time-dependent cytotoxicity
Carboxymethyl cellulose-AuNPs (CMC-AuNPs) Breast cancer induced apoptosis via caspase-8/9 activation and VEGFR-2 downregulation
Dunaliella salina-extract AuNPs Breast cancer Photoinduced AuNPs selectively killed cancer cells
Clerodendrum trichotomum (CTT)-AuNPs Breast cancer Reduced cell proliferation to 32.67% at
Marine algae-extract biosynthesized AuNPs Breast cancer showed strong anticancer activity (up to 92.13%) at 188 μg/mL
Equisetum diffusum-extract AuNPs Liver cancer Dose-dependent cytotoxicity; max 47.62% inhibition at 200 μg/mL; also shows antioxidant, antibacterial, and antidiabetic activity
AuNPs from Mimusops elengi in PVA/PCL nanofibers Skin cancer less toxic to normal fibroblasts (3T3); effective drug delivery system
M. elangi leaf-extract AuNPs in PVA/PCL nanofibers with curcumin Skin cancer Showed selective cytotoxicity via apoptosis; lower toxicity to normal cells; sustained drug release
Muntingia calabura (Mc)-AuNPs Laryngeal carcinoma AuNPs induced apoptosis, disrupted membrane, altered nuclear morphology, and caused G2 phase arrest
Cyclodextrin-covered AuNP Breast (MCF-7), Lung (A549) cancer Encapsulated b-lapachone; targeted delivery via EGFR; enhanced apoptosis
AuNPs with c-MWCNTs on GCE Cervical cancer Enabled sensitive detection of HeLa cells; used to evaluate anticancer activity of pinoresinol
Polygala elongata leaf-extract AuNPs Lung cancer Spherical AuNPs (10–20 nm) showed dose-dependent cytotoxicity with IC50
Acanthophora spicifera-mediated AuNP Colon cancer Spherical AuNPs (<20 nm) induced strong cytotoxicity (IC50 = 21.86 μg/mL) against HT-29 colon cancer cells, demonstrated antioxidant and antibacterial properties too
Ganoderma lucidum AuNPs–Doxorubicin conjugate Drug-resistant breast cancer enhanced drug delivery and gene (ABCB1) regulation
AuNPs synthesized from Exiguobacterium aestuarii Breast cancer Spherical AuNPs showed strong cytotoxic and apoptotic effects against cancer cells
Rauwolfia serpentina-mediated AuNPs Cervical cancer potential for novel targeted therapies
Euphorbia antiquorum-extract AuNPs Breast cancer Showed anticancer potential; active compounds (euphol, euphorbol) may inhibit VEGFR-2, ERK-2.
Mn(I)-carbonyl functionalized AuNPs Lung cancer Dual red light photoactivation induced CO release and singlet oxygen generation
T. capensis-extract AuNPs Breast cancer preventing the development and proliferation of human breast cancer cells
Kaempferol glycoside-mediated AuNPs Breast cancer showed mild to low cytotoxicity; strong antioxidant and catalytic activity
Sodium borohydride-synthesized AuNPs Lung cancer AuNP induced apoptosis and low toxicity to normal cells
AuNPs@ZnO (gold–zinc oxide hybrid) Breast, Liver showed anticancer potential but higher safety than ZnO alone
P. oleracea-extract AuNPs Breast, liver, cervical, colon showed stronger anticancer activity than crude extract via ROS-induced apoptosis
Bombax ceiba leaf-extract AuNPs Ovarian cancer Anisotropic AuNPs showed anticancer activity
Starch-reduced bimetallic AuNPs Melanoma 9.7 nm AuNPs showed dose-dependent cytotoxicity toward melanoma cells, while being cytocompatibility with normal human dermal fibroblasts (HDF)
Physalis minima-extract capped AuNPs Breast cancer Spherical AuNPs (22–32 nm) showed anticancer activity at 50 μg/mL; also exhibited strong antimicrobial and antioxidant effects
Levan-capped AuNPs loaded with DOX Breast cancer High drug encapsulation
HACD-AuNPs (β-CD–adamantane based supramolecular conjugates) General (e.g., DOX-tested tumor cells) Enabled targeted, pH-responsive delivery of multiple anticancer drugs with reduced toxicity and effective tumor inhibition
Achillea biebersteinii flower-extract AuNPs Testicular embryonic carcinoma apoptosis confirmed via gene expression analysis
Garcinia mangostana-derived Au–Ag core–shell NPs loaded with protocatechuic acid Colorectal cancer >80% inhibition at 15.63 μg/mL; higher selectivity over normal cells
β-lactoglobulin (BLG)-conjugated AuNPs Breast cancer Enhanced drug binding/stability with curcumin and gemcitabine
Cyanthillium cinereum leaf-extract AuNPs Lung cancer enhanced apoptosis compared to pure plant extract
Citrus macroptera fruit juice-derived AuNPs Liver, lung, breast cancer acted as antibiofilm agent
Pholiota adiposa polysaccharide (PAP-1a)–mediated AuNPs Hepatic carcinoma Enhanced immune regulation and antitumor effect
Enterococcus-mediated AuNPs Colorectal cancer Induced ROS, caspase-3 expression, mitochondrial dysfunction
M. tenacissima-extract AuNPs Lung cancer 50 nm AuNPs showed dose-dependent cytotoxicity, activated caspase and downregulated antiapoptotic proteins
Catharanthus roseus (CR)-AuNPs Cervical cancer Induced mitochondrial-mediated apoptosis via ROS
Curto-Cumin AuNP (CC-AuNP) biosynthesized using Nigella sativa and Curtobacterium proimmune Gastric adenocarcinoma Selectively cytotoxic to AGS cells; induced apoptosis via p53/Bax
Sasa borealis leaf-extract AuNPs Gastric cancer Oval/spherical AuNPs (10–30 nm) induced apoptosis in AGS cells
Plant-extract AuNPs (from Abutilon, Origanum, Euphorbia, Senna) Breast cancer Senna-based AuNPs most selective and potent
GMS-AuNPs and GMS-AuNPs@CS (Thyme-synthesized) Breast cancer Showed dose and time-dependent cytotoxicity; no toxicity to normal breast cells
Streptomyces flavolimosus-mediated AuNPs Breast (MCF-7), Cervical (HeLa), EAC (in vivo) reduced tumor growth in mice
Turmeric rhizome extract-based AuNPs Lung (A549) and Prostate (PC3) cancer phytochemical content influenced by altitude
A. muricata leaf-extract synthesized AuNPs Tongue squamous cell carcinoma (SCC-15) Induced apoptosis via upregulation of p53, Bax and downregulation of Bcl-2
Sonneratia alba fruit-extract AuNPs (SF-AuNPs) Lung cancer ∼30 nm AuNPs showed dose-dependent inhibition (up to 59.38% at 400 nM); exhibited antioxidant, anti-inflammatory, and anticancer properties
Camptothecin-loaded AuNPs Breast cancer apoptosis induction, antioxidant enzyme modulation, and PARP/Bax gene expression changes
Abies spectabilis-extract AuNPs Bladder cancer induced apoptosis via upregulation of Beclin-1
AuNPs@saponins niosomes (Sapindus mukorossi) Breast cancer Demonstrated anti-inflammatory potential with an IC50 of 30.08 μg/mL and showed anticancer activity in a dose-dependent manner.
AuNPs–PVP–Withaferin A–FA nanoconjugate Breast cancer pH-responsive and shows targeted delivery and anticancer efficacy
Marine bacteria (Vibrio alginolyticus)-synthesized AuNPs Colon carcinoma Induced apoptosis via nuclear condensation
Elephantopus scaber-extract AuNPs Breast (MCF-7), Lung (A-549), Oral (SCC-40), Colon (COLO-205) synergistic effect with Adriamycin enhanced anticancer activity across all tested lines
PSP-AuNPs (from Polygonatum polysaccharide) Hepatic carcinoma Showed strong anticancer effects in vivo with immune regulation

15.

15

Schematic mechanism of anticancer activity for AuNPs. Reprinted with permission from ref . Copyright 2021 Elsevier.

2.8.1. Enhanced Permeability and Retention Effect (EPR) and Tumor Targeting

Gold nanoparticles (AuNPs) show excellent stability, biocompatibility, and interesting optical properties, making them suitable as candidates for drug delivery to tumors. Their nanometer dimensions enable preferential tumor site accumulation through the EPR effect, wherein the leaky tumor vasculature traps nanoparticles while minimizing systemic clearance. Targeting ligand conjugation, e.g., antibodies, peptides, or folic acid, increases specificity and cancer cell uptake, enhancing therapeutic efficacy. Additionally, AuNPs act as photothermal agents, enabling targeted tumor ablation using plasmonic heating upon NIR illumination. These characteristics make AuNPs valuable nanomedicine tools for imaging and targeted therapy.

3.8.2. Imaging Application of AuNPs in Cancer

Gold nanoparticles possess unique optical properties, particularly surface plasmon resonance (SPR), which make them suitable for imaging applications in biomedical research. Their tenable size, shape, and surface chemistry allows for efficient functionalization with imaging ligands, enabling their use as contrast agents in molecular imaging and nuclear medicine (Table ). They can easily modify their surface with polymers, drugs, antibodies, and proteins, and they have become widely used as delivery vehicles. Their prolonged circulation time, enabling extended imaging and enhanced targeting efficiency make them suitable for CT imaging. AuNPs offer approximately 2.7 times greater imaging contrast than iodinated agents. Some traditional iodinated agents like iopamidol and iodixanol often cause allergic reactions and are unsuitable for patients with renal issues due to rapid clearance.

11. Summary of Imaging Applications of AuNPs Reported in the Literature.
imaging category AuNP formation method targeted area impact quantitative data limitation references
compound Tomography (CT) Citrate reduction followed by conjugation with 2-deoxy-d-glucose using mercaptosuccinic acid Human Lung A549 cells Enhanced tumor contrast on CT scans due to increased uptake of glucose- conjugated AuNPs CT intensity ∼900 HU with AuNP-DG (vs <300 HU with regular AuNPs) Uptake pathway not clearly understood; no in vivo testing yet
Photoacoustic Imaging (PAI) and OCT Femosecond laser ablation Chain like AuNP cluster assembled using CALNN + cysteamine, conjugated with PEG & RGD peptide Eye neovascularization in rabbit model (CNV) Strong dual imaging signal; excellent biodistribution and target specificity 176% increase in OCT signal Slightly complex synthesis Human translation still pending
MRI 24 nm sized AuNP were made via citrate reduction method Breast cancer cells Improved targeting and MRI contrast with low toxicity; promising for early tumor detection   Signal intensity lower than commercial MRI agents; purification step required
Photoacoustic Imaging & Photothermal Therapy (NIR-I and NIR-II) Seed mediated synthesis of nanorods and dumbbell-shaped, AuNPs, PEGylated for biocompatibility 4T1 Breast tumor in mice NIR-II window particles showed deeper tissue imaging and stronger heat generation for therapy AuNR1P, AuND1P, AuNR2P, and AuND2P exhibited high photothermal conversion efficiency values of 18.9%, 17.9%, 23.9%, and 25.3%, respectively. NIR-II AuNPs had slightly lower heating in tumors than NIR-I under same laser settings
Theranostics/Glioma Imaging & Therapy Various synthesis (e.g., Turkevich, Frens methods); functionalized with antibodies/peptides for brain targeting Glioblastoma (GBM) and CNS tumor AuNPs enable targeted therapy across blood-brain barrier, with potential for image-guided delivery No exact numeric data; qualitative confirmation of GNPs aiding brain-targeted delivery Difficulty in penetrating BBB; lack of standardized clinical formulations
Multimodal imaging (PAI,CT,MRI,PDT) AuNPs(spherical or rod shaped) engineered with antibodies/liposomes for selective targeting General cancer imaging (e.g., breast, glioma, colorectal) Demonstrated high specificity, stable imaging signals, and enhanced therapeutic payload delivery PAI, CT, MRI confirmed effective in vitro/in vivo models (no consolidated data set) Toxicity risks, particle aggregation, and immune clearance limit some applications
Fluorescence Hybrid QD@MSN-EPI-AuNPs, PEGylated with EpCAM aptamer Colorectal cancer (HT- 29 CELLS in mice) Targeted multimodal imaging and drug delivery Cumulative drug release (ph- 5.4): 76.33% in 6 days -
Emission peak shift from 610 to 630 nm
Photoacoustic Imaging Seedless synthesis of Au Nanostars with MOPS Buffer Deep tissue imaging High contrast signal 25 times better than spherical AuNPs AuNPs-MOPS produced 9.0 ± 0.0 PA units vs 4.7 ± 0.1 (EPPS), 4.0 ± 0.1 (HEPES), and ∼0.4 (spherical AuNPs); melanin coating enhanced signal 4.5 times Morphological heterogeneity may reduce reproducibility
PAI Guided therapy Janus chitosan-Au nanorods via counterion complexation + PEG Breast Cancer Synergistic gene/photothermal therapy; imaging + treatment Temperature elevation after 10 min NIR irradiation (808 nm): J-Au-CS = 30.4 °C vs Au NRs = 25.2 °C in cell Structural complexity may limit scalability
PAI Gold nanospheres coated with polydopamine-HA modifies Endometriosis lesions Targeted imaging of deep-seated lesions, enabled lesion volume tracking After irradiating with 808 nm laser for 10 min, temperature of 200 μg mL–1 PDA@HA nanoparticles increased by ≈22 °C, PA signal peaked at 8 h postinjection; lesion volume reduced by 66.93% (NAC group) Limited imaging depth due to NIR I absorption

3.8.3. Gold Nanoparticles in Drug Delivery

Gold NPs have gained significant attention for drug delivery due to their unique structural and functional properties. They have been explored fir drug delivery, particularly in cancer treatments and studies are being conducted on structure-efficacy relationships to optimize their design and application. Additionally, hybrid systems combining AuNPs with other therapeutic agents are being developed. A study conducted by Zazo et al. developed a gold nanoparticle-based system to improve the delivery of stavudine, an antiretroviral drug. The AuNPs used were citrate stabilized and 40 nm in size and prepared by incubating in stavudine solution for 24 h at room temperature. The formulation was tested for drug release at different pH levels and cellular uptake in human macrophages. In vivo studies in wistar rats showed that AuNPs increased stavudine accumulation in HIV (Human Immunodeficiency Virus) reservoirs like the liver, spleen, and macrophages. The liver showed a notable increase in MTR (mean residence time) from 1.28 to 5.67 h and partition coefficient was changed from 0.27 to 0.55. This system had enhanced biodistribution, prolonged drug residence and better targeting of latent HIV sites. No major limitations were directly reported, but the study emphasizes the need for further validation and clinical development. Sun et al. developed a thermosensitive nanoplatform combining AuNPs, hexanoyl glycol chitosan (HGC), a chemotherapeutic agent doxorubicin (DOX) and a photothermal; dye indocyanine green (ICG). AuNPs were synthesized via a citrate reduction method and them coated with HGC through electrostatic interaction, encapsulating DOX and ICG within the HGC shell. This assembly allowed drug release to be triggered by heat generated under 808 nm laser irradiation. It effectively targeted tumors and showed strong cytotoxicity in HeLa and SiHa cells and tumor-bearing mice. Advantages included enhanced therapeutic efficacy, targeted release, and minimal side effects. No major limitations were reported. Similarly, in the research conducted by Devi et al. AuNPs were synthesized using citrate and DOXmediated reduction, then functionalized with BP100 which is a cell penetrating peptide or RGD (targeting peptide). BP100@AuNPs-DOX showed high DOX loading (19.2 μM) with 96% encapsulation efficiency and up to 85% drug release at pH 4.5, better than RGD@AuNPs (70% release). BP100 systems showed superior cellular uptake and anticancer efficacy in HeLa cells. In HeLa cell assays, free DOX showed 32.1% killing, while DOX@AuNPs alone caused 27.2% death. Peptide-functionalized versions significantly improved efficacy-BP100@AuNPs-DOX and RGD@AuNPs-DOX killed 51.4 and 37.3% of cells, respectively. Overall, BP100@AuNPs-DOX exhibited the best anticancer activity, though high DOX concentrations reduced nanoparticle stability. In an experiment carried out by Zheng et al., 214 nm alginate-cysteine nanogels were embedded with 20 nm AuNPs. These DOX@ACA nanogels had 89.6% encapsulation efficiency and 3.7% drug loading. DOX release reached from 42.4 to 67.2% under 532 nm laser irradiation. DOX-AuNPs were highly responsive and effective but synthesis complexity and charge repulsion were the only noted limitations.

2.8.4. Limitations of AuNPs in Drug Delivery Applications

Although gold nanoparticles offer various benefits as drug delivery vehicles, their broader clinical application is primarily limited by concerns over safety. Factors including particle size, morphology, surface ligands, nuclei acid conjugates, dosage levels, and degradability influence their toxicity. To ensure reproducibility and consistent performance of gold nanoparticles (AuNPs) in drug delivery, maintaining uniformity in their size and shape is essential. Additionally, strategies for encapsulation, surface functionalization, and drug release must be specifically designed to match the physicochemical characteristics of the therapeutic agents being delivered. Release mechanism should be precisely controlled. A further challenge is ensuring the biological stability of AuNPs within physiological environments. Variations in temperature, pH, and the presence of ions or other biomolecules should not compromise the nanoparticle’s functionality or cause premature release or degradation of the loaded drug. Although these challenges demand rigorous design and testing, the unique properties of AuNPs continue to present significant opportunities in advancing drug delivery systems. While numerous studies have shown that AuNPs are generally safe, some have reported toxic effects. For example, a study conducted by Feng et al., involving bacteria such as Shewanella oneidensis (Gram-negative) and Bacillus subtilis (Gram-positive), gold nanoparticles coated with cationic or polyelectrolyte substances were found to exhibit higher toxicity compared to those functionalized with anionic ligands like 3-mercaptopropionic acid or the cationic ligand 3-mercaptopropylamine. Another study explored the use of gold nanoparticles embedded in or added to the soaking solution of contact lenses to enhance sustained release of Bimatoprost for glaucoma treatment. The researchers evaluated drug uptake, release duration, lens transparency, oxygen permeability, and protein adherence. However, certain limitations were identified. GNP- Laden contact lenses caused a high initial burst release of bimatoprost which may lead to side effects like eye redness. While drug release lasted up to 72 h, it dropped after 48 h, potentially below therapeutic levels. There is also concern about nanoparticle leakage, and long-term safety studies are still needed. The EPR effect was a pivotal discovery in nano-oncology, enabling passive targeting of nanoparticles to tumors. However, its clinical applicability has fallen short of expectations, as high accumulation of nanoconjugates in tumors via EPR alone is often inconsistent. , Consequently, the need for active targeting strategies remains critical to improve specificity and therapeutic efficacy in cancer drug delivery. ,

3.9. As Potent Quorum Quenchers

Gold nanoparticles (AuNPs) exhibit promising quorum quenching capabilities by disrupting bacterial quorum sensing (QS) pathways, which control virulence and biofilm formation (Table ). AuNPs inhibit QS by downregulating genes linked to bacterial communication, thereby reducing pathogenicity. These nanoparticles offer a potential alternative to antibiotics, especially against multidrug-resistant bacteria. Biosynthesized AuNPs, due to their eco-friendly nature and cost-effectiveness, are emerging as effective antimicrobial agents in combating bacterial infections and managing biofilms. These have shown promising potential as quorum quenchers, effectively inhibiting quorum sensing (QS) and virulence factors in Pseudomonas aeruginosa. Biosynthesized using Streptomyces isolate S91, these monodispersed AuNPs disrupted QS-related traits like pyocyanin, protease, and elastase production. The inhibitory effects were confirmed using RT-PCR, highlighting the potential of AuNPs as novel anti-QS agents for managing microbial resistance and chronic infections. Qais et al., showcased gold nanoparticles synthesized using Capsicum annuum extract demonstrated potent quorum-quenching capabilities against Pseudomonas aeruginosa PAO1 and Serratia marcescens MTCC 97. The AuNPs-CA effectively inhibited QS-regulated virulence factors, including pyocyanin, pyoverdin, elastase, and rhamnolipid production, along with biofilm formation. , Khosravi et al., study highlights the potential of green-synthesized AuNPs as biofilm, paving the way for new strategies to combat multidrug-resistant bacterial infections.

12. AuNPs as Potent Quorum Quenchers, Mechanisms, and Target Bacteria.

type of AuNPs/AuNCs functionalization/conjugation target bacteria mechanism/outcome references
Ultrasmall AuNCs (<2 nm) Acyl Homoserine Lactones (C-6, C-8, C-12) E. coli, C. sakazakii, P. aeruginosa Selective inhibition; C-8 AHL specific to C. sakazakii.
Chitosan AuNPs Chitosan, Antibiotics E. coli, S. aureus, P. aeruginosa pH-responsive Cur release; electrostatic bacterial targeting; ROS & heat generation kill
AuNPs (10–30 nm) N-acyl homoserine lactonase (AiiA) Proteus spp. (MDR) Inhibited QS signals, reduced biofilm formation, metabolic activity
Spherical AuNPs (168  ±  52 nm) Tetramethylpyrazine P. aeruginosa, S. aureus, S. mutans, K. pneumoniae, L. monocytogenes, E. coli Biofilm and virulence gene suppression; inhibits early/mature biofilms; reduces motility and toxins
SB-AuNPs (∼31.6 nm, aggregated spherical) Synthesized using quorum-quenching Salmonella bongori Pseudomonas fluorescens, Serratia marcescens Significant antibacterial, antibiofilm, and antiproliferative activity; AHL degradation
Biogenic AuNPs Ethnobotanical plant extracts Aeromonas hydrophila Biofilm and quorum sensing inhibition; enhanced antivirulence activity without resistance
AuNCs in Cs-GA copolymer composite Chitosan–Gum Acacia polymer matrix Pseudomonas aeruginosa Inhibits quorum sensing via LasR suppression; reduces biofilm, motility, protease; biocompatible

3.10. For Cosmeceutical Applications

Lately, Gold nanoparticles (AuNPs) are increasingly being incorporated in cosmeceuticals due to their antioxidant, anti-inflammatory, and antiaging properties. (Table and Figure ) outlines the various applications of AuNPs in cosmetics. AuNPs enhance skin permeability, deliver active ingredients, and offer resistance to environmental insult. AuNPs also promote collagen synthesis which reduces wrinkles and improves skin elasticity. However, further research is needed to know long-term safety and efficacy in cosmetic formulations. In cosmeceutical (that is cosmetic + pharmaceutical) applications, gold nanoparticles (AuNPs) offer promising potential for skin delivery systems. Curcumin which is a bioactive component of turmeric inhibits antioxidant and anti-inflammatory properties, with low bioavailability and instability. On encapsulating curcumin in chitosan-gold nanoparticles, the stability, absorption, and therapeutic activity are enhanced, and efficiency increases. Using chitosan derived from Oryctes rhinoceros beetle offers a green approach that converts pests into useful resources, gives remarkable stability, mild toxicity, and uniform nanosized particles, which are ideal for future cosmeceutical products. Nowadays, plant extract-mediated green synthesis of gold nanoparticles (AuNPs) is becoming increasingly popular because it is eco-friendly and does not utilize toxic chemicals. Despite limited studies, these are beneficial in antiaging, rejuvenation, and acne treatment creams and products. Due to their size and big surface-to-volume ratio, AuNPs penetrate skin layers with ease, facilitating the delivery of active components more efficient. Nevertheless, incorporating AuNPs in cosmeceuticals necessitates sensitive care of their environmental footprint, with a focus on green synthesis methods and lifecycle studies This approach ensures that the cosmetic industry can harness the unique benefits of AuNPs while prioritizing sustainability and minimizing ecological impact. Recent advancements include stimuli-responsive AuNP-collagen hydrogel nanoparticles (Au-CHPs), which enable controlled delivery of therapeutic proteins like fibroblast growth factor, superoxide dismutase, and epidermal growth factor. These Au-CHPs enhance wound healing, reduce oxidative stress, and promote skin regeneration, highlighting their potential for targeted, on-demand treatments in skincare. Singh et.al, in his research formulated a cosmeceutical peel-off mask by copolymerizing poly­(vinyl alcohol) (PVA) with sodium alginate, hydroxypropyl methylcellulose, or hydroxyethyl cellulose and incorporating silver nanoparticles (AgNPs). This approach was seen to enhance the antibacterial properties of the mask while maintaining its stability and biocompatibility. Similarly, gold nanoparticles (AuNPs) hold potential in cosmeceuticals due to their antioxidant, antiaging, and anti-inflammatory properties, enabling effective skin rejuvenation, wrinkle reduction, and enhanced product stability.

13. AuNPs in Cosmeceutical Applications.

methodology application key findings references
AuNPs conjugated with hyaluronic acid for topical application Targeted, pH-sensitive drug delivery system for cosmeceuticals and oral formulations. High encapsulation (94%) and sustained drug release; minimal release in acidic (gastric) and enhanced in basic (intestinal) pH.
AuNPs incorporated into botanical extracts antiaging skincare formulations to enhance cellular penetration and efficacy. improve antiaging activity of natural extracts, showing lower IC50 and higher inhibition than extracts alone.
AuNPs synthesized using marine algae and formulated into lotions As an antioxidant, and antibacterial cosmeceutical agent. Exhibited strong α-amylase & α-glucosidase inhibition, high antioxidant activity (DPPH & FRAP)
Green synthesis of AuNPs using extract of Hubertia ambavilla. Antiaging and dermoprotective cosmetic formulations. Nontoxic to human dermal fibroblasts; strong antioxidant and UVA-protective properties;
Green synthesis of AuNPs using Punica granatum juice (PGJ) Used as a natural antioxidant and SPF booster in sunscreen formulations. showed strong antioxidant activity, Improved SPF (3–18) depending on AuNP concentration.
Curcumin-loaded chitosan–AuNPs (CCG-NPs) synthesized using chitosan from Oryctes rhinoceros Stabilization and delivery of curcumin for cosmetic formulations using insect-derived chitosan. Spherical nanoparticles (∼128 nm), stable, mildly toxic (IC50 ∼ 58%), enhanced curcumin bioavailability and stability.
Green synthesis using Panax ginseng leaf extract. Antioxidant, moisturizing, and skin-whitening agent in cosmetics. Pg AuNPs showed dose-dependent antioxidant activity, moisture retention
Green synthesis of AuNPs using Helichrysum odoratissimum extract with/without gum arabic as stabilizer Antiacne cosmeceutical targeting Cutibacterium acnes adhesion HO-AuNPs significantly inhibited C. acnes adhesion, suggesting selective antiadhesion

16.

16

Cosmeceutical applications of AuNPs.

3.11. Gold Nanoparticle Application in the Treatment of Brain Dysfunction

Gold nanoparticles (AuNPs) are proven to be beneficial in treating brain dysfunctions due to their unique structure and properties, such as the ability to cross the blood-brain barrier (BBB) and deliver therapeutic agents directly to the brain. Some research and studies have shown that AuNPs are responsible for enhanced drug efficacy, reducing oxidative stress, and modulating neuroinflammation, offering neuroprotection in conditions like Alzheimer’s and Parkinson’s disease. However, further research is required to understand their long-term safety, biodistribution, and precise therapeutic mechanisms. Gold nanoparticles (AuNPs) also offer promising potential for treating neurological disorders due to their unique physicochemical properties. They can easily penetrate across the blood-brain barrier (BBB), facilitating drugs’ targeted delivery and minimizing off-target effects. In Alzheimer’s disease AuNPs inhibit amyloid-β aggregation, remove oxidative stress, and regulate neuroinflammation. In Parkinson’s disease (PD), they are neuroprotective through the removal of reactive oxygen species and inhibition of dopaminergic neuron loss. Nevertheless, challenges in toxicity, stability, and controlled release must be addressed for clinical use. Further research is necessary to optimize AuNPs for Safe and effective neurological interventions. In a recent study, AuNPs synthesized via microwave radiation and stabilized with dextrin were administered to diabetic rats. Diabetes can be mitigated by this treatment as it reduces oxidative stress, inflammation, and neurotransmitter imbalances. The higher dose (that is 2 mg/kg) was more effective than the lower dose (1 mg/kg), highlighting AuNPs’ potential in neuroprotection and managing diabetic brain dysfunctions. Gold nanoparticles (GNPs) linked with sodium diclofenac and/or soy lecithin were evaluated for safety and therapeutic potential in treating obesity-related inflammation. The 18 nm GNPs, administered intraperitoneally for 14 days, accumulated significantly in tissues without causing hepatic or renal toxicity. In obese mice, GNPs reduced food intake, and alleviated inflammation and oxidative stress, but did not reverse mitochondrial dysfunction. These findings suggest GNPs could be promising therapeutic agents, pending further safety assessments.

3.12. Photothermal Therapy

Gold nanoparticles inserted into gelatin hydrogels demonstrate strong photothermal properties under near-infrared laser light. These hydrogels enhance MC3T3-E1 preosteoblast, supporting bone regeneration. It produces heat in the localized area promoting cell growth and making this system promising in bone tissue engineering. They provide a minimally invasive and efficient way to improve bone healing through controlled thermal stimulation. AuNPs due to their adjustable optical properties and biocompatibility, are effectively used in plasmonic therapy. The radiation emitted from the laser converts light into localized heat, enabling the targeted destruction of cancer cells. Their surface can be modified for targeting tumors and reducing the damage to healthy tissues. Another study by Faid et al. presents hybrid chitosan-coated AuNPs as effective agents in photothermal therapy. They exhibit biocompatibility and stability, efficiently converting near-infrared light into heat to destroy cancer cells. The chitosan shells support in cellular uptake and prolongs circulation time. It shows significant potential in minimally invasive cancer treatment. A study by Frantellizzi et al. presented 99 mTc-labeled keratin-coated gold nanoparticles (Ker-AuNPs) designed for cancer photothermal therapy and imaging. They show biocompatibility and renal-clearance properties, modeled through a nephron-like system, ensuring minimal toxicity. This dual-function nano platform depicts integrated diagnosis and photothermal treatment in oncological applications. Similar study by Darvish et al. highlighted the use of keratin- coated AuNPs for photothermal therapy. The functionalized AuNPs serve as promising agents for cancer therapy and therapeutic effects. The study investigates anisotropic gold nanoparticles stabilized with choline carboxylic acid ionic liquids for photothermal therapy. They show excellent thermal conversion under infrared light, enabling targeted cancer cell ablation. Their unique structure enhances the stability, biocompatibility, and efficiency. They are effective agents in noninvasive cancer treatments through localized heat generation. Green synthesis of gold nanoparticles for photothermal therapy in combination with chemotherapy. They generate heat to kill cancer cells, enhance drug uptake and therapeutic efficiency. The biocompatible approach reduces side effects and improves the cancer treatment. Another study discussed the nanocellulose-based codelivery system that enhances photothermal therapy. Gold nanoparticles (AuNPs) combined with curcumin are integrated with carboxymethylated cellulose nanofibrils, enhancing the stability, solubility, and release rate. The system boosts photothermal effects under near-infrared irradiation, leading to efficient cancer cell destruction. This method offers a biocompatible and synergistic treatment combining thermal and chemotherapeutic effects with minimal side effects.

3.13. Antibacterial Properties

4-mercaptobenzoic acid (MBA)-functionalized gold nanoparticles (AuNPs) have demonstrated potent antibacterial activity (Table ), particularly against multidrug-resistant (MDR) S. aureus and S. epidermidis. Similarly, 4,6-diamino-2-pyrimidine-thiol (DAPT) functionalized gold nanoparticles embedded in a silk fibroin (SF) membrane showed good antibacterial activity against drug-resistant and drug-sensitive strains of E. coli. Controlled release of the DAPT-AuNPs was provided by the hydrophilic nature of the SF membrane. In vivo experiments with SD rats revealed quick healing of the wound in 3 μg/cm2-treated groups compared with control groups, or groups treated with plain SF membranes or gauze. Besides surface changes, particle shape is also an important determinant of the antibacterial activity of AuNPs. Irregular AuNPsrod, star, peanut, and porous sphereswere found to cause membrane integrity loss of E. coli and P. aeruginosa. Irregular shapes facilitate interaction with bacterial surfaces, enhance the generation of reactive oxygen species (ROS), and enhance antibiofilm efficacy. All these mechanisms contribute to efficient inhibition of bacteria and biofilm formation inhibition.

14. Antibacterial Potential of AuNPs.

type of AuNPs targeted bacteria observed effects references
Ellagic Acid-Modified AuNPs (EA-AuNPs) Multidrug-resistant ESKAPE pathogens High bactericidal efficacy; mitigated inflammatory responses
Aminophenol-Modified AuNPs (AP_Au NPs) Multidrug-resistant bacteria Damaged bacterial cell walls; bound to 16S rRNA, blocking protein synthesis; effective in vivo against abdominal infections.
Thioproline-Modified AuNPs E. coli Potent antibacterial activity; balance between monodispersity and aggregation crucial.
D-Maltose-Capped Au Nanoclusters with Thiourea (AuNC-Mal/TU) Multidrug-resistant Pseudomonas aeruginosa Effective at low MIC; inhibited thioredoxin reductase; interfered with Cu+ regulation; depleted ATP.
Graphene Oxide–Gold Nanoparticles (GO–Au NPs) S. aureus, E. coli Significant antibacterial activity; effective against both Gram-positive and Gram-negative bacteria.
Vitex negundo Leaf Extract Synthesized AuNPs Salmonella typhi, Micrococcus luteus Exhibited antibacterial activity; zones of inhibition observed; potential for biomedical applications.
Virola oleifera-Synthesized AuNPs S. aureus, Pseudomonas aeruginosa Significant antibacterial activity against both strains;
Phospholipid-Stabilized Gold Nanorods E. coli, S. aureus Effective photothermal antibacterial activity; stability and low toxicity
Codariocalyx motorius-Synthesized AuNPs Various bacterial and fungal pathogens Exhibited antibacterial and antifungal activities
Metallic-Core AuNPs S. aureus, Klebsiella pneumoniae, Streptococcus pneumoniae, E. coli, Brucella abortus, Mycobacterium bovis Displayed good antibacterial activity against all tested pathogens; potential alternative to traditional antibiotics.

3.14. As Antiviral Agents

Antiviral drugs and agents are in great demand throughout the world. Each year millions of infections occur due to the attack of these viruses thus causing infections. Metal nanoparticles have the intrinsic property of antiviral activity which provides a potential solution to mitigate these infections. AuNPs can attach to viral particles, thereby preventing their interaction with cellular or viral receptors and inhibiting the initiation of the viral replication cycle. AuNPs adhere to the cell surface and alter the membrane potential, thereby blocking the viruses from entering the cell. This antiviral action of AuNPs (Table ) has been attributed to several mechanisms, including the prevention of virus attachment and entry into host cells, interference with plasma membrane binding, inactivation of viral particles prior to entry, and interaction with double-stranded DNA. Gold nanoparticles inhibit a broad spectrum of viruses, including HIV, influenza, and herpes simplex virus due to their antiviral properties. In fact, some research showcased the antiviral potential of gold nanoparticles (AuNPs) against SARS-CoV-2, focusing on their ability to disrupt viral envelopes and prevent viral entry. Ayurvedic metal nanoparticles, such as Swarna Bhasma have unique properties like anti-inflammatory, immunomodulatory, and antiviral effects, making them promising agents for COVID-19 treatment. Furthermore, study is required to authenticate their therapeutic potential against SARS-CoV-2 and other viral infections leading to diseases. Glaucium flavum leaf extract was known to produce stable, spherical AuNPs with a mean size of 32 nm and low aggregation. It was synthesized in a green way and characterization techniques like FTIR, and GC-MS confirmed their stability and bioactivity. The potential of functionalized gold nanoparticles (AuNPs) as an effective antiviral agent against herpes simplex virus (HSV) by inhibiting viral entry and replication was observed by Ayipo et al., in his work. These nanoparticles undergo surface modification of AuNPs to enhance their binding affinity to viral proteins, allowing targeted. These findings highlight the potential of AuNPs in developing advanced antiviral therapies. Similarly, sulfonic group-modified gold nanoparticles (MDS_AuNPs) possess broad-spectrum via antiviral properties but lose efficacy in high-protein environments. To address this, sulfonic mixed-charge modified gold nanoparticles (MC_AuNPs) were developed by introducing positively charged ligands. MC_AuNPs retained antiviral activity even in a 10 mg/mL protein solution, unlike MDS_AuNPs, which failed in 1 mg/mL solutions. Chaika et al., in their work showcased that these gold nanoparticles (AuNPs) exhibit potent antiviral activity against adenovirus and H1N1 influenza virus, with 5 nm size indicating stronger virucidal effects compared to 20 nm AuNPs. The smaller nanoparticles were observed to disrupt viral structures within 2 h, while larger ones made shape changes. AuNPs are universal virucidal agents with low cytotoxicity and minimal reactive oxygen species (ROS) generation due to this physical adsorption mechanism.

15. AuNPs as Antiviral Agents, Target Virus, Cells, and Formulations.

AuNP formulation target virus synthesis method size of AuNP (nm) targeted cells/organisms references
AuNPs functionalized with peptide, Mes, Mus, Ot SARS-CoV-2 Molecular dynamics 2 Spike protein RBD; blocks ACE2 binding
PEG-AuNPs PEG-AuNPs Turkevich chemical reduction 2–10 HIV particles, HIV-infected CD4+ T cells
HIV
AuNPs with sulfonated glucose/lactose ligands DENV-2 Ligand coating + docking study 4–5 Hepatocytes; DENV-2 envelope protein
DNA-conjugated AuNP network Human RSV Self-assembly on cell membrane 13 Inhibits viral attachment, entry, budding, and spread
AuNPs using garlic extract (AuNPs-As) Measles virus (MeV) Green synthesis with Allium sativa 6 Vero cells; blocks viral particles (virucidal)
AuNPs–PNA conjugate BVDV (HCV model) PNA loading on AuNPs 13 ± 1 nm Infected cells; targets viral RNA (5′-UTR)
Nonfunctionalized AuNPs HSV-1 Turkevich method 10 or 16 Smaller AuNPs more effective Vero cells; HSV-1 envelope
Citrated and PEG-dithiol functionalized AuNPs BVDV Citrate reduction; PEGylation 12 nm (citrated), 15 nm (PEGylated) MDBK cells; inhibits viral attachment
HA–AuNP/IFNα complex HCV Chemical binding (thiolated HA) + physical loading of IFNα 22.16 Liver tissue
AuNPs/LDHs (e.g., MgFeLDH) Hepatitis B Virus (HBV) Self-assembly using LDH AuNPs ∼ 3.5 nm on LDH ∼ 150 nm HepG2.2.215 cells
Lactoferrin-modified AuNPs HSV-2 Chemical + Lactoferrin coating varied Human keratinocytes
AuNPs coated with poly(styrenesulfonate) (PSS) Broad-spectrum viruses RAFT polymerization + gold salt reduction 3.1 Viral particles

3.15. Gold Nanoparticles in Chemical and Biological Sensing

Gold nanoparticles (AuNPs) are now being widely used in biological and chemical sensing due to their unmatched optical and electronic properties, which enable easy detection of analytes (Table ) Figure . AuNP-based sensors control the electron transport mechanisms and surface plasmon for electrical and optical readouts. Environmental monitoring, medical diagnostic and real-time detection in complex matrices due to its rapid response times, selectivity, and potential for miniaturization. Gold nanoparticles play a crucial role in biosensors due to their unique properties. Surface Plasmon Resonance (SPR) enables them to exhibit collective electron oscillations when exposed to specific wavelengths of light, allowing for the detection of subtle refractive index changes in the surrounding medium. This characteristic is essential for high-sensitivity biosensing. Additionally, their high surface area to volume ratio facilitates the attachment of numerous biomolecules, improving sensor performance. Fungal-mediated synthesis techniques offer precise control over nanoparticle size and shape, further enhancing surface area and making them highly effective for sensing applications. Moreover, Au NPs are biocompatible and nontoxic allowing functionalization with biomolecules like antibodies, DNA, and peptides without affecting their biological compatibility and making them ideal material for medical and environmental sensing.

16. Chemical and Biological Sensing Applications of AuNPs.

Type of AuNPs (with reason) functionalization target analyte sensing mechanism references
Spherical AuNPs (citrate-capped)Uniform size distribution and surface plasmon resonance (SPR) optimized for visible light detection. Unmodified Water Pollutants (Hg2+, Pb2+) Colorimetric detection via SPR shift; aggregation causes color change
AuNPs conjugated with anti-SARS-CoV-2 spike antibodies via PEG linker (Ab-PEG-AuNPs)for specific viral protein recognition and enhanced signal transduction Antibody (antispike) functionalized using EDC/NHS chemistry and PEG linker SARS-CoV-2 virus (Spike protein) Microwave sensing enhancement through specific antigen–antibody interaction; label-free detection
Gold Nanoclusters (2 nm, Ligand-Protected) - Atomically precise, fluorescence in NIR region. Ionic Liquid Functionalization Ascorbic Acid Aggregation-induced quenching for fluorescence detection
Bimetallic gold nanostars coated with silver (BGNS-Ag)sharp spikes and Ag shell offer high SERS enhancement and reproducibility Surfactant-free (unmodified) Methylene blue, Thiram (pesticide) SERS-based detection with ultrahigh sensitivity and minimal sample prep
AuNPs functionalized with mixed PEG-thiol ligandsstudy highlights limitations in uniform grafting of mixed layers. Mixed thiol-PEG ligands (methoxy, carboxylate, alkyne ends) Ligand grafting efficiency Quantitative 1H NMR analysis to assess ligand density on AuNP surface
Colloidal Gold NanoparticlesEnhanced stability in physiological conditions. Bioactive agents Biomarkers Electrochemical detection via enhanced electrocatalytic activity
Plasmonic AuNPs Unmodified Energetic charge carriers Time-resolved emission upconversion microscopy via plasmon decay and carrier cooling

17.

17

Summary of different types of gold nanoparticle biosensors. Reproduced from ref . Copyright 2018, with permission of Elsevier.

3.16. Gold Nanoparticle-Based Plasmonic Organic Solar Cells

Plasmonic solar cells are an advanced type of photovoltaic technology that utilize the unique optical properties of metal nanoparticles (usually gold, silver, or aluminum) to enhance light absorption and increase solar cell efficiency (Table ). The key concept behind plasmonic solar cells is the Surface plasmon resonance (SPR), which refers to the collective oscillation of conduction electrons at the surface of metal nanoparticles when excited by incident light. Studies have shown that the incorporation of plasmonic nanostructures in solar cells significantly enhances their power conversion efficiency (PCE). The improvement in PCE largely depends on the type of solar cell, the material used, the shape and size of plasmonic nanoparticles, and their position within the device structure. Gold nanoparticles, despite having lower heat-generation capability than silver or copper, are preferred due to their chemical stability and resistance to oxidation. Heat generated through plasmonic effects is transferred from the nanoparticle to the surrounding medium, affecting the local temperature and thus enabling various applications such as photothermal therapy, catalysis, and sensing.

17. AuNPs Applications in Solar Cells and Their Impact on Performance.

Sr. No. size shape solar cell composition impact on performance references
1 41 nm Spherical Organic (PBDTTT-CF:PC70BM) Increased efficiency from 6.67 to 7.86%
2   Spherical Perovskite (CdS-grated) Improved Jsc, PCE, VOC value and FF
3 70 nm Spherical Schottky (silicon based) 47% increase in J sc with significant heat generation
4 Core–Shell (Au@TiO2, Au@SiO2) Spherical perovskite Increased optical absorption, improved charge separation, and enhanced stability.
5 10–150 nm Spherical Perovskite-Organic Tandem Mitigates optical losses, improves electron–hole recombination, and enhances current gain by >1.5 mA/cm2, achieving 25.34% efficiency.
6 Fe3O4@Au Core–Shell   Hybrid organic (magnetoplasmonic) 8.3% increase in PCE
7 16 nm Spherical Polymer (P3HT:PCBM) 16% PCE Improvement via plasmonic enhancement of light absorption
8 51 nm (AuMPA) and 12 nm (for AuOA,) Spherical Hybrid bulk heterojunction Improved J sc, PCE of the device
9 87 spherical Polymer bulk heterojunction 87 NM SIZED NPS due to the balanced contribution of near- and far-field plasmonic effects, have improved vertical coverage and better interfacial properties. Thus, improve efficiency
10   Truncated polyhedral Organic (P3HT:PC71BM) Higher PCE as compared to nanocubes due to better plasmonic coupling
11 Au and Ag NPs (1 wt%) spherical Organic(P3HT:PCBM) PCE increased was observed from 2.11% to 2.55% (Au) and 2.23% (Ag) due to enhanced light absorption.
12   Decahedral Polymer (P3HT:PC60BM, PBDT-TS1:PC70BM) Improved light response across broadband wavelengths, improving PCE to 10.29%
13 10–20 nm spherical inverted bulk heterojunction organic solar cell (PTB7-Th:PC71BM) 24% efficiency improvement, PCE reaching to 11.8%, due to plasmonic enhancement in ZnO electron transport layer.

3.17. In Veterinary Medicine

Gold nanoparticles have gained attention in recent years for their multifunctional role in nanomedicine, particularly in drug delivery and theranostics. Their unique ability to be tailored in different size and shapes influences their efficiency in imaging, drug encapsulation, and targeted therapeutic applications. One of the notable uses of AuNPs in veterinary medicine is in rapid disease detection. Moongkardi et al. developed an immunochromatographic assay for detecting bacterial infections such as Salmonella enterica serovars Typhimurium and Enteritidis, in poultry. These assays provide a quick and reliable means of identifying pathogens and can help to improve food safety and animal health. Similarly, AuNPs-biosensors have been employed for the detection of haptoglobin in mastitic milk, thus helping in early detection of subclinical mastitis in dairy cows. AuNPs also play a crucial role in antimicrobial applications. Kumar et al. developed gold nanoparticle-based immunogens for the detection of colistin, a critical antibiotic in veterinary medicine in chicken liver samples. Additionally, AuNPs have been investigated for extending the shelf life of chilled minced meat by inhibiting bacterial growth, particularly against E. coli and Salmonella. In therapeutics, AuNPs have been studied for targeted drug delivery and immunomodulation. Loghmani et al. developed betaine-conjugated AuNPs in a murine model of heatstroke, where they significantly reduced inflammation and oxidative stress by modulating cytokine levels and enhancing immune function Moreover, they are also integrated into regenerative medicine strategies, such as stem cell therapy tracking and tissue repair, demonstrating their potential for veterinary application. The role of AuNPs in role regeneration and dental applications has also been explored, with studies investigating their ability to enhance osseointegration of dental implants in rabbit models. Furthermore, AuNP-based hydrogels have been developed for use in regenerative medicine, providing a biocompatible scaffold for tissue engineering applications.

3.18. AuNPs in Dental Biomaterials

Modern healthcare prioritizes increasing patient survival rates and improving quality of life, with a key focus on developing implantable materials that can mimic natural biological functions. Currently a range of materials such as metals, ceramics, carbon nanostructures, and polymers are widely used in orthopedic and oncological surgeries. However, these materials face challenges such as limited strength, poor biointegration, metal ion diffusion, and toxicity from degradation byproducts. To mitigate these complications, researchers are developing bioactive surface treatments that provide antimicrobial protection Several studies of use for titanium and titanium-based alloys are being reported. Noble metals like platinum, palladium, silver, and gold have gained attention due to their exceptional biocompatibility, resistance to corrosion, and antibacterial properties. Coating implant surfaces with ultrathin layers or nanoparticles of these metals has demonstrated benefits in promoting bone integration and reducing inflammation Due to its biocompatibility, and chemical and corrosion resistance in biological environments, metallic gold and its alloys are being used for implants and surgeries. There are two primary forms of gold known for their antibacterial properties. The first is nanoporous gold (NPG), which features a highly porous structure at the nanometre scale. This unique morphology enhances its surface area, enabling effective interaction with bacterial cells. Studies have demonstrated that gold nanoparticles exhibit antimicrobial effects against E. coli and Staphylococcus epidermidis, making it a promising material for medical and biomedical applications. Solanki et al. synthesized AuNPs using Triphala extract which were found to be biocompatible and possess strong antimicrobial properties, making them a potential alternative for oral care products. Researchers evaluated the cytotoxicity of these nanoparticles and an AuNP-based mouthwash using the Artemia salina (brine shrimp) assay. The mouthwash exhibited mild toxicity at higher concentrations (40–50 μL), with a mortality rate of 46.6%. At lower concentrations (20–30 μL), the mouthwash was found to be safe and effective, suggesting its suitability for use in orthodontic patients. Another study suggested that gold nanoparticles (AuNPs) enhance the biocompatibility, proliferation, and antioxidant properties of dental biomaterials, making them suitable for tissue regeneration and restorative applications Biz et al. combined gold nanoparticles with poly­(l-lysine) (AuNP-PLL) and introduced into dental pulp stem cells (DPSC) which resulted in high cellular uptake without compromising cell viability or inducing apoptosis. This highlights AuNPs’ potential for imaging and monitoring stem cells in regenerative endodontics Dharman et al., synthesized gold nanoparticles using curcumin which demonstrated excellent antimicrobial and anti-inflammatory properties and can be used for treating oral infections and mucosal lesion Dalavi et al., synthesized spherical AuNPs (70 nm) using a microwave irradiation method which exhibited strong antioxidant activity and cytocompatibility with IMR-32 cells and thus can be used for cosmeceuticals and pharmaceutical application. Dentures, which serve as artificial, nonshedding surfaces in the oral cavity, are primarily composed of poly­(methyl methacrylate) (PMMA). AuNPs were synthesized via ultrasonic spray pyrolysis and incorporated into PMMA poly­(methyl methacrylate) to improve its physical and mechanical properties, additionally enhancing its antimicrobial properties. The modified material exhibited similar density and microhardness to conventional PMMA but demonstrated a significant reduction in monomicrobial biofilms of Candida albicans, Streptococcus mitis, S. aureus, and E. coli Another study incorporating gold nps with PMMA resulted in enhanced antifungal activity and improve oral hygiene for denture wearers. These nanocomposites demonstrated significant antifungal activity against Candida albicans at concentrations above 2.0%, with minimal ion release.

3.19. In Active Food Packaging

Shelf life of food is one of the major global concerns for food loss which increases the importance of advanced packaging solutions. Nanotechnology, particularly metal-based nanoparticles, offers a promising approach to extending shelf life while addressing environmental issues. Nanotechnology in the food industry is predominantly used for processing, packaging, and detecting contaminants like toxins, microbes, pesticides, and food adulteration. Nanoparticles aid in enhancing the taste, smell, texture, look, and shelf life of food products, offering improved quality and safety throughout the supply chain. Green synthesis of gold nanoparticles (AuNPs) is gaining attention for its efficiency, reduced biohazards, and enhanced antimicrobial and antioxidant properties which will be beneficial for food packaging (Table ). These have remarkable antioxidant, antibacterial, antifungal, anticancer, and barrier properties along with being inert, nontoxic, hypoallergenic nature and biocompatible to humans. AuNPs can be incorporated into smart packaging systems to improve food safety, quality monitoring, and shelf life. However, factors such as nanoparticle concentration, food type, polymer composition, and storage conditions influence their effectiveness and migration into food, necessitating careful evaluation for safe application in packaging. One of the unique properties of AuNPs particularly are surface plasmon resonance (SPR), peroxidase-like activity and their influence on sensing performance. SPR-based sensors include aggregation, antiaggregation, etching, and growth-based methods, each with varying sensitivity and selectivity. Peroxidase-based (nanozyme) sensors offer a higher sensitivity but involve complex protocols. Smaller AuNPs (<10 nm) increase the catalytic activity, while larger ones (>10 nm) improve SPR-based detection. Despite strong potential, colorimetric sensors are less explored than electrochemical ones for inorganic ion detection. AuNP-based colorimetric sensors offer a simple, fast, and cost-effective method for food safety monitoring without any need for complex equipment. However, challenges remain, including interference from food matrices and the need for specific ligand modifications to enhance selectivity and sensitivity. Future advancements should focus on improving analyte extraction, developing highly specific aptamers, and incorporating signal amplification strategies to enhance detection in real-world food safety applications. Biogenic NPs improves the food shelf life and can serve as biosensors for real-time quality assessment. While promising, challenges remain regarding nanoparticle migration into food and potential toxicity, requiring further research to ensure safety in food-related applications. Sreelakshmi et al. explored the use of chitosan, derived from shrimp waste, as a reducing and capping agent in synthesizing gold nanoparticles (AuNPs) for smart packaging applications in the food and pharmaceutical industries. The reduction time for gold atoms varies depending on the chitosan type, ranging from 6 to 15 min. Higher concentrations of chitosan give rise to smaller, more uniform AuNPs. When exposed to freezing conditions (−18 °C ± 1 °C), the ruby red color of AuNPs shifted to bluish or colorless, which indicates the temperature fluctuation. This visible change confirms the potential of chitosan-based AuNPs as indicators for distinguishing between fresh and frozen products. Alghamdi et al. had also done a study which focuses on developing bionanocomposite films by incorporating gold nanoparticles (AuNPs) into a chitosan (CS) and polyacrylamide (PAM) polymer blend by application of solution casting method. AuNPs were synthesized using Chenopodium murale leaf extract. XRD analysis revealed a crystallinity reduction and an increase in amorphousness by addition of AuNPs. TEM images showed nearly spherical AuNPs, while FTIR spectra confirmed strong interactions between the nanoparticles and the polymer matrix, which was indicated by shifts and intensity changes in functional group bands. UV–vis analysis showed a decrease in both direct and indirect bandgap energies, along with an increase in Urbach energy, reflecting enhanced optical properties. TGA results showed improved thermal stability in Au-CS/PAM composites in comparison to the pure blend. Mechanical testing showed enhanced Young’s modulus, tensile strength, and elongation at break. Additionally, Au-CS/PAM nanocomposites exhibited impressive antimicrobial and antioxidant activity, pointing to their usefulness as active food packaging materials and in optoelectronic applications. Choudhary et al. conducted a study, where poly­(vinyl alcohol) (PVA) composite films incorporating gold nanoparticles (AuNPs) and graphene oxide (GO) was developed and cross-linked using glyoxal or glutaraldehyde (GA). FTIR analysis confirmed effective cross-linking through reduced hydroxyl group transmittance. The addition of AuNPs and GO enhanced mechanical and physical attributes such as tensile strength, Young’s modulus, water vapor transmission rate (WVTR), and water solubility. WVTR tests revealed that nanofillers contributed to reduced permeability by forming a complex, tortuous structure. SEM images showed compact pore morphology in cross-linked films in comparison to pure PVA. Antibacterial activity was observed in both AuNPs- and GO-based composites against E. coli, with PVA-glyoxal-AuNPs films showing a major inhibition zone, indicating superior antimicrobial performance. Besides that, this film extended the shelf life of bananas more effectively than others, confirming its potential for food packaging. In conclusion, the PVA-glyoxal-AuNPs composite exhibited promising structural, antimicrobial, and preservative properties for advanced food packaging applications. Mehmood et al. highlighted the potential of gold nanoparticles (AuNPs) conjugated with gallic acid (GA) as an effective strategy to combat reactive oxygen species (ROS), a major factor in packaged food spoilage. Amine-stabilized AuNPs were synthesized and functionalized with GA from Caesalpinia pulcherrima extract, reducing toxicity and improving antioxidant efficiency. GA-AuNPs exhibited a strong free radical scavenging ability, shown by cyclic voltammetry, with a low IC50 (4 × 10–9 g/mL) and high antioxidant coefficient. Even though antimicrobial activity was minimal, GA-AuNPs achieved 94.1% DPPH scavenging, outperforming GA and its derivatives. The study confirms GA-AuNPs to be a safe, multifunctional material for extending food shelf life and serving as a nontoxic antioxidant carrier in food packaging applications. Yang et al. developed a novel ratiometric electrochemical sensor for the simultaneous detection of two endocrine-disrupting compounds (EDCs) as 17β-estradiol (E2) and bisphenol S (BPS), using a composite of gold nanoparticles (AuNPs) and MIL-101­(Fe). Traditional electrochemical sensors have been commonly facing challenges in detecting EDCs in food due to poor reproducibility. To tackle this problem, the sensor utilized the Fe signal inherent to MIL-101­(Fe) as a built-in internal reference, replacing the need for external electroactive tags which are commonly used in ratiometric sensing. AuNPs were electrodeposited onto MIL-101­(Fe), which resulted in a composite material with enhanced electrochemical properties. The stability of the Fe reference signal and the oxidation responses of E2 and BPS has remarkably boosted due to synergistic effect between AuNPs and MIL-101­(Fe).The developed sensor has a wide linear detection range from 0.039 to 7.80 μM and achieved low detection limits of 9.8 nM for E2 and 11.2 nM for BPS. Application in milk sample analysis confirmed the sensor’s excellent reproducibility, sensitivity, stability, and practical performance. This work provides a promising direction for constructing ratiometric electrochemical sensors with the use of electroactive MOFs for rapid and accurate EDC detection in food safety monitoring.

18. Key Findings of AuNPs in Active Food Packaging.

Sr. No. application area key findings references
1 Antibacterial Activities AuNPs exhibit significant antibacterial, antifungal, and antibiofilm properties.
Synergistic effects with chitosan, APTMS, and other compounds enhance antimicrobial activity.
Effective against Gram-negative and Gram-positive bacteria.
Shape and concentration-dependent antimicrobial properties.
2 Barrier Properties AuNPs improve water vapor, gas (CO2, O2), and light barrier properties. ,,
Enhanced molecular structure tortuosity reduces permeability.
Optimal concentration is crucial to avoid agglomeration and maintain barrier efficiency.
3 Antioxidant Properties AuNPs interact with free radicals, showing dose-dependent antioxidant potential.
Green-synthesized AuNPs (e.g., using onion peel extract) exhibit notable radical scavenging capacity.
High surface-to-volume ratio enhances antioxidant activity.
4 Biosensing AuNPs are used in colorimetric sensors for detecting food spoilage and contamination.
High sensitivity and rapid response due to large surface-to-volume ratio.
Effective in detecting volatile biogenic markers and heavy metal ions.
5 General Benefits AuNPs improve mechanical, thermal, and surface properties of packaging materials. ,
Enhance shelf life and food safety.
Migration behavior of AuNPs into food requires careful regulation due to potential toxicity concerns.

3.20. Agricultural Applications

The synthesis of nanoparticles (NPs) has garnered significant attention due to their distinctive properties, which make them valuable for a wide range of applications, including composite fibers, biosensors, cryogenic superconducting materials, cosmetics, and electronic components. However, in light of climate change and the depletion of natural resources, there is a growing emphasis on sustainable methods for producing gold nanoparticles (AuNPs) and silver nanoparticles (AgNPs). One promising approach involves using plant extracts, particularly agricultural waste, as a green and eco-friendly alternative. This method aligns with sustainable development goals in agro-industrial practices. Given that plants serve as the foundation for this green synthesis, the resulting NPs are not only environmentally friendly but also exhibit low toxicity, making them suitable for various agricultural applicationsfrom soil treatment to food chain integration. , In June 2009, the Food and Agricultural Organization (FAO) and the World Health Organization (WHO) highlighted the potential of nanotechnology in food and agriculture. Their joint initiative identified several key areas for innovation, including nanostructured ingredients, nanosized biofortification, food packaging, nanocoating, and nanofiltration. NPs can boost productivity of specific plant tissues or structures by beneficial genes, delivering nutrients, or organic compounds. This capability positions NPs as advanced nanodelivery systems, specifically for enhancing crop nutrition and agricultural efficiency. In agriculture, AuNPs have been extensively studied for their direct applications, which include improving seed germination, promoting root growth, and understanding plant responses to metal NPs, also cellular oxidative stress and cytotoxicity. Nanofertilizers and nanopesticides are also being developed by direct use of metal Nanoparticles. Indirect applications of nanoparticles (NPs), using their antimicrobial properties, are primarily centered on advancements in food packaging. These advancements have been extensively imparted in the agricultural industry, with AuNP based products typically incorporating particles ranging from 100 to 250 nm in size, this size range enhances their water solubility and overall effectiveness. In short, the embedding of NPs into agriculture and food systems represents a novel and transformative approach, offering solutions that are both innovative and aligned with sustainability principles.

3.20.1. In Water Treatment

Water is a vital part of the ecosystem and its treatment highlights the need. Several techniques have been developed for this purpose. Agriculture accounts for almost 70% of the world’s renewable water resources which makes it important to consider for wastewater treatment of agricultural water. In order to enhance sustainable agricultural practices, it is necessary to assess the impact of agro nanobiotechnology on water conservation and its quality. To enhance sustainability and efficiency Nanotechnology is gaining attention in agriculture. A notable approach is the development of gold nanoparticles through environmentally friendly biogenic synthesis using natural sources like plants, fungi, and bacteria. These nanoparticles have strong antibacterial and antifungal properties, making them useful in safeguarding crops from harmful pathogens. Moreover, they can aid in pesticide detection and water purification, promoting safer and more effective farming methods. By incorporating nanotechnology into agricultural practices, researchers strive to improve crop protection and optimize resource use while maintaining environmental responsibility. Water pollution caused by nitrophenol, a nitrogen-containing pollutant, creates significant environmental concerns. A novel approach using thiourea-treated gold nanoparticles (AuNPs) to create nanoporous films via filtration was explored for efficient catalytic degradation of nitrophenol into the less toxic aminophenol. The study found that AuNP films treated with 20 μg/mL thiourea and 1000 mM NaBH4 facilitated rapid conversion within 150 s. These films demonstrated remarkable structural stability and retained 90% catalytic efficiency after seven cycles. The stable mesoporous AuNPs film results in a cost-effective and sustainable solution for industrial wastewater treatment, making it a promising strategy for environmental remediation. A study by Francis et al. utilized a rapid microwave-assisted method to synthesize gold and silver nanoparticles using Mussaenda glabrata leaf extract as a reducing and stabilizing agent. Characterization techniques, including UV–vis, FT-IR, XRD, TEM, and AFM, confirmed the nanoparticles’ FCC crystal structure. Both nanoparticles exhibited strong antioxidant activity and antimicrobial properties against pathogens like Pseudomonas aeruginosa and E. coli. They showed effective degradation of pollutants such as rhodamine B, methyl orange, and 4-nitrophenol, making them promising catalysts for wastewater purification.

3.20.2. Nanofertilizers, Nanopesticides, and Nanoherbicides

Agriculture plays a vital role worldwide by ensuring food security and economic stability. Traditional ways for boosting crops include the usage of chemical fertilizers, insecticides, and herbicides. However, excessive use of these agrochemicals has led to soil degradation, loss of biodiversity, and environmental concerns. Nanotechnology approaches are used to enhance crop production, improve food security, and develop pest- and drought-resistant crops. Nanoengineered materials improve nutrient absorption, enable rapid disease detection, and function as nanofertilizers and nanopesticides, boosting productivity while minimizing soil and water contamination. These materials also protect against microbial diseases and pests which reduces chemical use and nutrient loss. Additionally, nanotechnology aids in soil quality monitoring, ensuring optimal crop yields. Ongoing research in agricultural nanotechnology is being used to enhance food quality, safety, and efficiency, supporting sustainable agricultural practices for a growing global population. Its applications have been used across multiple areas, such as precision farming, food preservation, and plant protection, due to the distinctive properties of nanomaterials. These materials are known for their controlled-release capabilities, targeted action on specific sites, and large surface area, making them highly efficient in agricultural use. Nanofertilizers, nano herbicides, and nano pesticides enhance plant growth, optimizing nutrient absorption, and provide effective pest and weed control, ultimately leading to higher yields with very less environmental impact. Over the past decade, nanotechnology in agriculture, specifically nanofertilizers and nanopesticides, has gained attention for its potential to transform farming. This analysis has reviewed scientific, regulatory, and commercial progress, highlighting emerging products, differing sector perceptions, and the challenge of risk-benefit assessment. It focuses on the need for improved formulations, clearer definitions, and smarter, sustainable agrochemical development.

3.21. Textiles Industry Applications

Nanotechnology has a vital role in expanding textile applications across various fields, such as protection, fashion, sports, healthcare, the military. By modifying textiles at the nanoscale with functional nanomaterials, it is possible to introduce new features while maintaining their comfort and usability. The textile industry has greatly advanced with the introduction of new applications, especially when combined with nanomaterials. Different types of textilessuch as woven, knitted, and nonwoven fabrics, as well as fibers, yarns, threads, nanofibers, scaffolds, and membranesare now used in high-tech and smart applications. Adding gold nanoparticles (AuNPs) to textiles brings exciting new features, including unique colors, improved filtration, antimicrobial properties, conductivity, UV protection, sensory functions, and catalytic abilities, as explored in this section.

3.21.1. Chemical Reduction Method without Pretreatments on Fabrics

Chemical reduction is one of the most used methods for production of textile-fictionalized gold nanoparticles (AuNPs), due to its effectiveness and simplicity. However, there is a growing shift toward greener and more cost-effective alternatives. Traditionally, researchers follow a two-step functionalization approach, where AuNPs are first synthesized and then applied to fabrics. Alternatively, some opt for an in situ approach, where synthesis and deposition occur in a single step. In the two-step method, sodium borohydride (NaBH4) and sodium citrate used as common reducing agents. Various deposition techniques have been explored, such as dropwise deposition, exhaustion, padding, impregnation, and printing with AuNP dispersions. For example, Chan et al. (2016) synthesized AuNPs using chloroauric acid (HAuCl4) as the precursor, NaBH4 as the reducing agent, and sodium citrate as a capping agent. They applied the nanoparticles to cotton, silk, and wool fabrics using the dropwise deposition method. Zheng et al. synthesized Gold nanoparticles (AuNPs) via citrate reduction, were immobilized onto chitosan-treated soybean knitted fabric by use of the exhaustion method. Moderately polydisperse AuNPs had an average size of ∼35 nm. Successful immobilization was confirmed through spectrophotometric reflectance, X-ray photoelectron spectroscopy (XPS), and Fourier-transform infrared spectroscopy (FTIR). XPS analysis showed strong AuNP-chitosan binding. The coated fabrics exhibited enhanced thermal stability, ultraviolet protection (UPF 50+), and antimicrobial properties, effectively reducing S. aureus (99.94%) and E. coli (96.26%) adhesion. The coating also displayed durability, withstanding five washing cycles with minimal AuNP loss with additional benefit of coloration. XPS analysis suggested AuNPs bound to chitosan in a pure metallic state, though potential antimicrobial mechanisms involving oxidized Au species and reactive oxygen species require further study. These multifunctional fabrics show promise for biomedical applications due to their UV shielding, optical properties, and antimicrobial effectiveness. Shanmugasundaram and Ramkumar has utilized keratin with silver and gold nanoparticles to develop antibacterial wound-healing materials. Human hair, a major waste from barbershops, contains keratin (a biocompatible protein) with wound-healing and antibacterial properties. The nanoparticles, synthesized by chemical reduction, were characterized using UV–visible spectroscopy, particle size, and ζ-potential analysis. Silver and gold nanoparticles formed at 420 and 479 nm with average sizes of 71.8 and 14.59 nm, respectively, and negative ζ-potential values of −18.9 and −3.2 mV. FTIR confirmed the presence of keratin and nanoparticles in coated cotton fabrics. SEM images showed uniform, high-density coatings, while EDX confirmed high oxygen and carbon content. The coated fabrics exhibited excellent physical properties, such as air permeability, moisture content, and water absorbency. Additionally, they demonstrated superior antibacterial activity against burn wound bacteria, making them promising for biomedical wound-healing applications. Lin et al. synthesized Nylon fabrics through heat treatment with citrate assistance by in situ technique. The synthesized AuNPs impart bright colors to the fabrics due to their localized surface plasmon resonance (LSPR) properties. The optical characteristics were analyzed using color strength (K/S) curves, while SEM was used to observe surface conditions. The synthesis process was influenced by pH, with acidic conditions favoring AuNP formation. The treated fabrics exhibited excellent color fastness to washing and rubbing. Additionally, the coloration process significantly enhanced the UV-blocking properties of Nylon fabrics. This study provides both aesthetic and functional benefits, offering a promising approach for textile coloration and UV protection at the same time.

3.21.2. Chemical Reduction Method with Pretreatments on Fabrics

Radić et al. conducted study that evaluated two ambient air plasma treatments including volume dielectric barrier discharge (DBD) and diffuse coplanar surface barrier discharge (DCSBD) to enhance gold nanoparticle (AuNPs) deposition on polypropylene (PP) nonwovens. Plasma treatments used to improve surface wettability and sorption, increasing AuNP loading from 17 mg/kg (untreated) to up to 62 mg/kg (DBD-treated). DBD enhances the surface roughness, while DCSBD induces notable chemical changes. Antibacterial tests showed effective activity against S. aureus and E. coli, with higher sensitivity observed in S. aureus. Surprisingly, rinsing enhances the antibacterial properties without reducing AuNP content, due to altered nanoparticle clustering. These findings suggest that AuNPs-loaded plasma-treated PP nonwovens are promising for reusable antibacterial materials, with the choice of plasma method influencing surface and functional properties. Ikegami et al. developed a novel filter-type Au/ZrO2 catalyst using PET nonwoven fabric as a lightweight, flexible support, offering advantages over conventional catalyst forms. ZrO2 particles were deposited with a silane agent to create a thin, fish-scale-like layer, followed by gold nanoparticle deposition. This catalyst effectively removed 1000 ppm of CO (83% conversion in 20 min) and 140 ppm formaldehyde (90% removal, 68% oxidized to CO2 in 90 min) at room temperature. Nearly 100% removal of 0.5 ppm formaldehyde was found in tests simulating indoor conditions for up to 136 h, demonstrating strong performance and long-term effectiveness for air purification applications. A flexible and sensitive surface-enhanced Raman spectroscopy (SERS) substrate was developed by depositing uniform Au nanoparticles onto polydopamine-coated cotton fabrics using Ag nanoparticles as catalytic hotspots. The in situ reduction process produced a dense, even layer of AuNPs which was confirmed by SEM, XRD, and XPS analyses. The substrate, CF/Ag/PDA/Au, showed strong and reproducible SERS signals using 4-MBA as a probe, detecting concentrations as low as 10–9 M. It also successfully detected carbaryl pesticide residues on cucumbers reduced to 10–6 M, below regulatory limits. This work highlights the potential of CF/Ag/PDA/Au as a flexible, durable, and effective SERS platform for real-world food safety monitoring. Hence Gold Nanoparticles can be seen as a greener and sustainable approach in the textile industry.

3.21.3. Electrochemical Synthesis Method for Textile Applications

In this there is integration of gold nanoparticles (AuNPs) into textiles to create antimicrobial fabrics, emphasizing eco-friendly and effective synthesis and deposition methods. Two main electrochemical approaches have been identified: A two-step process involving separate AuNP synthesis and deposition, and a more efficient one-step in situ synthesis directly on textiles. While the in situ method saves time but still relies on chemical agents for gold reduction. Innovative techniques like plasma treatment and thermal activation using silk fibers have shown promise in enhancing nanoparticle adhesion and reducing chemical usage. These electrochemical strategies have the potential for scalable, low-toxicity fabrication of antimicrobial textiles, especially for biomedical and hygiene applications. However, more research is needed to optimize these methods and evaluate long-term safety and effectiveness. Over the past two decades, gold nanomaterials (AuNMs) have gained remarkable attention due to their unique catalytic properties at the nanoscale. Electrodeposition is a highly controllable synthesis method, commonly used to create gold nanoparticles, nanoclusters, and nanowires. Enhancing both activity and stability by designing monodisperse nanoclusters, multimetallic nanoparticles, and tailoring surface-support interactions is an important area in this. AuNMs have illustrated versatility in chemical, photochemical, and electrochemical catalysis. Their biocompatibility also finds usage in biomedical, such as drug delivery and photodynamic cancer therapy. These advancements position AuNMs as highly promising materials for diverse applications across scientific field.

3.22. AuNPs as Wearable Sensor

A Study by Zhang et al. showed the development of self-healing and conductive elastomer for wearable sensors. The material consists of poly­(dimethylsiloxane) (PDMS) and gold nanoparticles (AuNPs) by utilizing sulfur–gold (S–Au) interactions. When AuNPs are exposed to near-infrared (NIR) light, the material repairs itself by achieving healing efficiency of 92%. The elastomers exhibit electrical conductivity and strain sensitivity, making it suitable for applications such as monitoring human joint movement and muscle activity in health monitoring and soft robotics. Another study depicted a wearable strain sensor based on the ligand-exchanged AuNPs for detecting human motion. It consists of thin layer of 9 nm gold nanoparticles (AuNPs) deposited on a poly­(dimethylsiloxane) (PDMS) substrate, offering high flexibility and sensitivity capturing small movements of fingers and wrist. With the help of the ligand exchange the conductivity is enhanced, making it well suited for biomedical applications. Another study by Khorablou et al. depicts highly sensitive and flexible sensor which detects methadone. It is built using gold nanoparticles (AuNPs) and polythiophene on a carbon cloth platform, improving sensitivity and enhancing electron transfer. It showed successful application in human blood and urine samples which helps to demonstrate real-time drug detection. For instance, another study discusses about the advancement of wearable sensor for detecting cardiovascular disease (CVD). AuNPs enhance the sensors’ conductivity, sensitivity, and biocompatibility allowing to monitor vital signs like pulse waves, heart sounds, and electrocardiogram (ECG) signals. Their integration into flexible, lightweight material supports real-time health tracking and early cardiovascular disease detection. They offer potential applications in continuous, noninvasive diagnostics and personalized healthcare. A study by Chen et al. demonstrates a wearable electrochemical biosensor utilizing gold nanoparticles (AuNPs) for in situ pesticide detection on crops. It is built on a flexible fiber membrane, a three-electrode system modified with acetylcholinesterase (AchE) and reduced graphene oxide (rGO) to enhance performance. It detects methyl parathion with a low detection limit of 0.48 ppb, providing a rapid and nondestructive method for agricultural monitoring. The development of wearable strain sensors using cross-linked gold nanoparticles (AuNPs) via contact printing method. These sensors are integrated into flexible polyimide (PI) and poly­(dimethylsiloxane) (PDMS) substrates. They demonstrate high strain sensitivity, durability over 10,000 usage cycles, and a rapid response making them well-suitable for healthcare applications. Another study depicted implantable sensors using AuNPs present hydrogel-embedded sensors for continuous biomarker monitoring. They utilize plasmon resonance shifts to detect analyte concentrations through the skin. The tests on anesthetized rats successfully detected kanamycin levels, depicting long-term stability and integration into tissue. A study by Chen et al. demonstrated a wearable glucose sensor designed for continuous monitoring through human sweat, incorporating gold nanoparticles (AuNPs) with aminated multiwalled carbon nanotubes (AMWCNTs) and cross-linked with XSBR and PEDOT: PSS. Integrated onto screen-printed electrodes, it shows high sensitivity, flexibility, and stability making it a potential tool for health tracking and diabetes management. The wearable capacitive sensor designed to monitor leaf moisture was created by depositing gold nanoparticles (AuNPs) onto a poly­(ethylene terephthalate) (PET) membrane using magnetron sputtering, ensuring stable conductivity and adaptability to plant surfaces. The change in capacitance provides a noninvasive method for assessing plant hydration. It has promising applications in agriculture and environmental monitoring. For instance, a study by Wang et al. introduced a sweatband sensor capable of real-time sodium ion, incorporating an all-solid-state ion-selective electrode (ISE) and a reference electrode (RE) by ensuring high sensitivity and stability. It is fabricated via electrodeposition on a flexible substrate, allowing continuous monitoring of hydration levels and electrolyte balance, showing potential applications in personalized healthcare, sports science, and medical diagnostics. A similar study by Dau et al. developed a wearable colorimetric sensor for glucose detection in sweat, integrated with an automated microfluidic chip. It features a glass fiber-based electrode enhanced with gold nanoparticles (AuNPs), improving color stability and sensitivity. This device is designed for continuous health track monitoring, diabetes management, and personalized healthcare applications.

3.23. Gold Nanoparticles in Nanoelectronics

Gold nanoparticles (AuNPs) are used in biomedicine due to their high stability, biocompatibility, and flexible properties (Table ). They play an essential role in drug delivery, biosensing, imaging, and photothermal therapy. Their surface modification enables targeted therapeutic applications by reducing its side effects. Advancements in nanotechnology are enhancing their efficiency for clinical use, medicine, and disease treatment. The plasmonic gold nanoparticles (AuNps) focus on the optical properties, method of synthesis and various biomedical uses. Their plasmonic properties enhance diagnostic techniques such as surface-enhanced Raman spectroscopy (SERS). A study by Oladipo et al. discusses about the biosynthesis of gold nanoparticles (AuNPs) using Datura stramonium seed extract. It is characterized via UV–vis, FTIR, SEM, and EDX, and depicted antifungal, antioxidant, anticoagulant, and thrombolytic activities. They are effective in inhibiting fungal growth, neutralizing free radicals, prevented blood coagulation and clot dissolution. It shows potential applications in biomedicine, particularly for antimicrobial and cardiovascular treatments. For instance, study by Clarance et al. depicted the synthesis of gold nanoparticles (AuNPs) using endophytic fungus Fusarium solani. AuNPs were characterized via various techniques and potent anticancer properties against breast cancer (MCF-7) and cervical (HeLa) cells. The biosynthesized AuNPs were found to trigger apoptosis, block cell proliferation, and induce cell cycle arrest. It shows potential chemotherapeutic agents. Another study by Das et al. explores the synthesis of gold nanoparticles (AuNPs) using Amaranthus plant extract. They exhibit various biomedical applications such as drug delivery, bioimaging, and antimicrobial activity. It shows antioxidant and anticancer properties making is well suited for therapeutic use. The research shows potential applications in nanomedicine, enhancing biocompatible nanomaterials for advanced medical treatments. Gold nanoparticles (AuNPs) have significant applications in nanoelectronics due to their excellent conductivity, stability, and customizable optical properties. They are used in sensors, transistors, and memory devices, allowing miniaturization and enhanced performance of electronic components. Functionalized nanoparticles with organic molecules further improve the efficiency of device and their self-assembly properties, making them ideal for nanocircuit fabrication. GNPs contribute to flexible electronics and molecular-scale computing, forwarding next-generation nanoelectronics technologies. A study by Babajani et al. focuses on the controlled stabilization in heterometallic nanogaps. They use various fabrication techniques, surface modifications, and their influence on electrical characteristics. It shows advanced nanoscale electronics, enabling advancements in molecular circuits, sensors, and quantum devices by distinctive optical and electronic characteristics. Another study investigates the electroless deposition (ELD) of silver thin films on SiO2/Si surfaces activated by AuNPs. It depicted successful Ag film growth through distinct stages by improving their electrical and optical properties. AuNPs provide a cost-effective, result in rougher films as compared to traditional palladium activation. It improves film quality by using smaller AuNPs to refine surface morphology and conductivity. A similar study by Ruiz et al. demonstrates the use of DNA origami as a template, leveraging the self-assembling properties of DNA developing nanoscale and circular metallic structures. They enable the precise placement of gold nanoparticles, crucial for miniaturized electronic devices. The fabrication process using atomic force microscopy and gel electrophoresis, depicts the potential of DNA origami for future nanoelectronics application.

19. Gold Nanoparticles in Biomedical Applications.

Sr. No. cell type shape size synthesis applications references
1 HeLa and MCF-7 cells Needle and Flower like Size-40–45 nm Green synthesis using Fusarium solani fungus Shows anticancer by inducing apoptysis and cell cycle arrest, potential chemotherapeutic effect
2 Fibroblast (3T3 cells) Spherical Size varies- (AuNps-38 nm and keratin albumin-591 nm) AuNps synthesized using Keratin and albumin Used in drug delivery, Biocompatibility and potential therapeutic applications
3 Gloeocapsa sp. and Anabaena cylindrica Spherical, cubical, and triangular Size ranging from 2–100 nm Synthesized via Extracellular or Intracellular using enzymes and proteins Includes drug delivery, biosensing and anticancer treatment
4 HepG2 and A549 cells Spherical Size range between 2–10 nm Synthesis using Asprgillus niger culture supernatant, reducing chloroauric acid and silver nitrate at pH 8, 100 °C Highlights the potential in thrombosis treatment and cancer therapy
5 A549 and HepG2 cells Cubic 16.51 nm Green synthesis using Morchella esculenta hot water extract at room temperature Biocompatible agents for therapeutic applications, including microbial inhibition and cancer treatment
6 Hela and A549 cells quasi-spherical 49.72 ± 1.2 nm Green synthesis using Euphrasia officianalis leaf extract and cholorauric acid reducion Showed significant anticancer activity against HeLa cells (cervical cancer cells) but no cytotoxicity effect toward A549 (lung cancer cells)
7 Not specified Cubic range between 20–50 nm Green bioreduction using Chrysopogon zizanioides aqueous extract and chloroauric acid Shows significant antibacterial, antioxidant and cytotoxic activities
8 HT-29 cells Spherical to oval Gold nanoparticles size between (7.2–46.9 nm) Synthesized using Caulerpa racemosa extract through a bioreduction process Shows anticancer activity against HT-29 cells and antibacterial effect against A. veronii and S. agalactiae
9 Not specified(In vivo study in rats) Spherical Gold nanoparticles(12 nm) Synthesized using the citrate reduction method and conjugated with peptides, labeled with [18F]-fluorobenzoate Potential for imaging (PET), drug delivery and Alzheimer’s disease treatment
10 HepG2 cells Spherical, Triangular, and hexaganol size range ∼31 ± 1.6 nm Synthesized using Eclipta Prostrata leaf extract and HAucl4 reduction Potential use in cancer therapy, drug delivery and antimicrobial treatments
11 Not specified Anisotropic ranges between 10–18 nm Rapid green synthesis using Adiantum philippense L. frond extract, reduction of tetrachloroauric acid at pH11 Potential for cancer therapy and antimicrobial treatments.
12 MCF-7 and NIH3T3 cells Spherical 35 ± 16 nm Synthesized using the Citrate reduction and precipitation method Potential applications in drug delivery, cancer therapy, and biomedical imaging
13 Not specified Spherical Not specified Synthesized using Colloidal methods for stable and functionalized structures applications in medical diagnostics and imaging, MRI contrast enhancement
14 Not explicit but potentially in cancer cells and inflammatory cells Spherical 20–30 nm Synthesized using Capsicum annum fruit extract potential use in cancer treatment, oxidative stress management, and inflammation reduction,
15 Various human and bacterial cells Shapes include spherical, rod-shaped, and star-shaped nanoparticles Sizes typically range from 1–100 nm Green synthesis using plant extracts, microbial synthesis, and chemical reduction methods Used in drug delivery, cancer therapy, bioimaging, biosensors, wound healing, and antimicrobial treatments.

4. Toxicity of AuNPs

Gold nanoparticles showed potential use in biomedical, but their toxicity can vary depending on the size, shape, surface charge, and functionalization (Table and Figure ). Smaller particles penetrate easily leading to oxidative stress, inflammation, or genetic damage. Surface modification improves the toxicity of AuNPs. Gold nanoparticles synthesized using melatonin reduced the toxicity and enhanced biocompatibility. Melatonin served as both a reducing agent and stabilizing agent, producing uniform nanoparticles with antioxidant properties. They show minimal cytotoxicity in cell viability assays by interacting safely with biological environments. This approach can reduce the negative impacts of AuNPs, depicting safer use in medical applications like drug delivery and imaging. Another study examines the size and surface chemistry of gold nanoparticles influencing their impact on neurons. The particles also build up in cell structures, disrupting their function. They highlight the importance of nanoparticles for safe use in treatments involving the nervous system. Biosynthesized gold nanoparticles using plant extracts exhibit reduced cytotoxicity due to their natural capping agents. Biocompatibility and eco-friendly synthesis make them suitable for biomedical applications. Tables , , and depicts toxicity of AuNPs tested on various mice models, Zebrafish models and on various cell line.

20. Factors Influencing the Toxicity of Gold Nanoparticles (AuNPs) as Described in the Literature.

factor description & influence on toxicity
1. Assay Interference In vitro assays (MTT, LDH, CFE, etc.) may give inconsistent results due to differences in AuNP size, shape, dose, surface chemistry, and cell types, limiting reliable toxicity evaluation.
2. Size Smaller AuNPs (<5 nm) often show higher toxicity due to larger surface area-to-volume ratio, increasing interaction with biomolecules; however, effects may vary between in vitro and in vivo models.
3. Shape Spherical AuNPs tend to be more toxic than nanorods, often due to residual synthesis agents like CTAB.
Spherical AuNPs are more commonly used due to ease of synthesis and lower toxicity.
4. Surface Chemistry Functionalization (e.g., PEG, peptides, , nucleic acids) affects stability, targeting, and uptake.
Surface coatings reduce aggregation and can minimize toxicity or alter cellular responses and biodistribution.
5. Surface Charge Positively charged AuNPs show higher cellular uptake and toxicity due to interactions with negatively charged membranes,
Neutral or negatively charged particles are generally less toxic.
6. Aggregation State Aggregation increases toxicity; influenced by surface charge, size, and coating agents.
Preventing aggregation (e.g., via PEG, BSA) reduces cytotoxicity. ,
7. Interactions with Media/Fluids Proteins and salts in biological fluids form a protein corona, altering AuNPs’ charge, size, and behavior. ,
This may reduce targeting ability, trigger immune responses, or influence biodistribution. ,
8. Sex Differences In vivo toxicity may differ between sexes due to hormonal, anatomical, and metabolic differences.
Some studies reported higher liver toxicity in male mice.
9. Dose Toxicity is dose-dependent; higher doses may not reflect clinical relevance. Low-dose models offer better human relevance, and effects vary with route and exposure duration.
10. Route of Administration IV injection generally causes lower toxicity; oral and intraperitoneal routes may cause more damage due to mucosal absorption and systemic exposure.
11. Uptake & Biodistribution Uptake depends on particle size, shape, charge, and coating. ,
Smaller particles show wider tissue distribution; liver and spleen show highest accumulation, which may or may not correlate with toxicity.
12. Clearance & Excretion Particles <6 nm are cleared renally; larger particles are cleared hepatobiliary or retained in RES (e.g., liver, spleen).
Surface chemistry (e.g., PEGylation) can improve clearance and reduce long-term toxicity.

18.

18

Toxicity mechanism of gold nanoparticle. Reprinted with permission from ref . Copyright Elsevier B.V.

21. Toxicity of AuNPs in Mice.

Sr. No. organism method of synthesis particle size & shape dosage & route exposure duration capping/reducing agent ζ-potential observed toxic effect outcome references
1 Male albino rats Citrate reduction Gold nanoparticles (AuNPs): 13 ± 4 nm, spherical 570 μg/kg/day, intraperitoneal 28 days Citrate (reducing + capping agent)   Decreased body and testicular weight, reduced testosterone, sperm count/motility, abnormal sperm morphology, histopathological changes (seminiferous tubule disruption, Sertoli and Leydig cell degeneration, mitochondrial damage) Reversible reproductive toxicity (partial recovery at 30 days, near-complete at 60 days)
2 Mice Citrate & NaBH4 reduction Gold nanoparticles (AuNPs): 3–100 nm, spherical 8 mg/kg/week, intraperitoneal 21 days Sodium citrate   Sizes 8–37 nm caused systemic toxicity (fatigue, weight loss, fur changes, spinal deformities, high mortality); histopathology revealed liver Kupffer cell proliferation, lung damage, splenic disorganization Toxicity was size-dependent; particles <8 or >37 nm showed no adverse effects. Immunogenic peptide coating reduced toxicity
3 Mice Citrate reduction Gold nanoparticles (AuNPs): 2, 40, 100 nm, spherical 5 doses, intratracheal 3 weeks Citrate Negative No significant toxicity; 2 nm particles showed minor liver accumulation; larger particles remained lung-localized; macrophage uptake was prominent Translocation was size-dependent; nanoparticles were largely retained in lung macrophages with negligible systemic toxicity
4 BALB/c mice, F344 rats Commercial Gold nanoparticles (AuNPs): 15 nm, spherical 1000 mg/kg, intravenous, single dose Acute Citrate & PEG Negative Mice: liver granulomas, IL-18 elevation, no lethality; Rats: spleen accumulation, fecal excretion, partial mortality Demonstrated species-specific biodistribution and immunogenicity; inflammatory but nonlethal response in mice, higher toxicity in rats
5 Wistar rats Citrate reduction Gold nanoparticles (AuNPs): 10, 30, 60 nm, spherical Intravenous (dose not specified) - Citrate   Smaller AuNPs (10 nm) caused higher DNA damage, oxidative stress, nuclear localization; accumulation noted in liver, spleen, kidney, and intestines; inflammation and tissue injury increased with smaller size Demonstrated size-dependent toxicity; 10 nm particles induced stronger genotoxic and inflammatory effects compared to larger ones
6 Rats Citrate reduction Gold nanoparticles (AuNPs): 20 nm, spherical 0.01 mg/kg, intravenous 2 months Trisodium citrate   No overt toxicity; however, liver and spleen showed gene expression changes related to metabolism, detoxification, and immune function; minimal testis accumulation; no brain distribution Demonstrates long-term biodistribution with subcellular, organ-specific effects without clinical toxicity
7 C57BL6/J mice Biosynthesis (P. pterocarpum leaf extract) Gold (b-Au-PP): 54.2 nm, spherical (hydrodynamic) Daily, intraperitoneal (7 days) 7 days P. pterocarpum (biocapping/reducing) –21.1 mV No significant hematological, biochemical, or histological toxicity; nanoparticles were stable and well-tolerated Demonstrated excellent short-term biocompatibility; biosynthesized AuNPs show potential for biomedical applications
8 C57/Bl6 mice Antibody conjugation Gold (AuNPs-Cetuximab) Core: 4–5 nm; Total: ∼26 nm, spherical 90 μg Au/mouse, intravenous, single dose Up to 6 months Poly allylamine, Cetuximab –7.04 mV No acute toxicity in major organs up to 4 weeks; kidney casts and splenic apoptosis observed at 6 months Good short-term safety profile; potential long-term organ-specific effects warrant further evaluation
9 Wistar rats Citrate reduction Gold (AuNPs) 10, 50, 100, 250 nm, spherical 1 mL/rat, intravenous, single dose 24 h Citrate   No overt systemic toxicity; 10 nm particles distributed to multiple organs including brain; larger particles mostly confined to liver, spleen, and blood Biodistribution was size-dependent; smaller particles exhibited broader organ penetration without acute toxicity
10 Mice, fibroblast cells Chemical or green synthesis Gold nanoparticles (AuNPs): 25–30 nm, spherical Systemic or dermal   Phytochemicals (capping/reducing agents)   ROS production, inflammation, apoptosis, DNA damage, epigenetic alterations, cellular dysfunction Small size, crystalline structure, and surface charge influenced toxicity; surface modification recommended to reduce toxicity
11 Male Wistar rats Not specified; characterized by TEM and UV–vis Gold nanoparticles (AuNPs) 5–50 nm, spherical 25–250 mg/kg, intramuscular or intravenous       Reduced testosterone, altered liver enzymes (ALT, AST, ALP), altered kidney markers (urea, creatinine), histological changes in testes, oxidative stress, hormonal disruption, cellular damage Tissue accumulation and hormonal imbalance highlight reproductive and hepatic toxicity concerns; size and surface chemistry influenced outcomes
12 Mice Turkevich method BSA-coated AuNPs ∼ 20 nm, spherical 1 mg/kg, intravenous (IV) Up to 120 days Bovine Serum Albumin (BSA)   Accumulation in liver, spleen, kidneys; kidney inflammation; liver/spleen fibrosis; fibronectin expression; inflammatory gene upregulation Long-term retention led to subchronic toxicity; fibrotic changes in organs noted.
13 Pc-Au NCs Study (Rats) Green synthesis (potato extract) Gold nanoparticles: Spherical, 12 nm (AuNPs), 20 nm (Pc-Au NCs) 10 μg/kg/day, IP 3 weeks Phytochemicals (potato extract), Phthalocyanine –22.7 mV (AuNPs), –19 mV (Pc-Au NCs) No toxicity; anti-inflammatory and protective effects Safe profile at low dose
14 Male BALB/c mice Citrate reduction Gold Nanoparticles: Spherical; 10 nm (GnP10), 50 nm (GnP50), 100 nm (GnP100) 4 mg/kg IV (tail vein), alone and with drugs (cisplatin, paraquat, 5-ASA) 24 h Citric acid Negative (due to citrate coating) GnP10: Strong nephrotoxicity when coadministered with drugs (↑IL-6, BUN, Cr); GnP50: mild effects with 5-ASA; GnP100: no toxicity, even with drugs Toxicity was size-dependent: smaller particles (GnP10) showed strong interaction and toxicity; larger ones (GnP100) were biocompatible even with toxic drugs
15 Male ICR mice Turkevich method Gold nanoparticles (AuNPs): 13.5 nm, spherical 137.5–2200 μg/kg; oral, intraperitoneal, and intravenous (tail vein) routes 14–28 days Citrate Not numerically stated; negative due to citrate Low doses: no toxicity; High doses: reduced body weight, RBC count, spleen index (especially oral/IP). IV had least toxicity Toxicity was dose- and route-dependent; tail vein was safest, supporting its use for biomedical applications
16 Male albino rats Commercial (citrate-stabilized) Gold nanoparticles (GNPs): 10 nm, spherical 20 μg/kg, intraperitoneal injection 7 days Citrate Negative (citrate buffer) Oxidative stress, lowered antioxidants (SOD, GST), decreased neurotransmitters, brain inflammation and damage Sulforaphane reversed toxic effects by boosting Nrf2 activity, antioxidants, and neural function
17 Swiss albino mice Sodium borohydride reduction Thiol-PEG capped gold nanoparticles ∼4.5 nm, spherical IV (dose not specified), single/acute exposure Not specified (short-term) Thiol-functionalized triethylene glycol Not specified; improved stability No cytotoxicity (in vitro), no histological or biochemical toxicity; distributed in liver, kidney, tumors High safety; favorable biodistribution and nontoxic even in tumor-bearing mice at high concentrations
18 Male C57/BL6 mice Citrate reduction (HAuCl4) Gold nanoparticles (GNPs) ∼12.5  ±  1.7 nm, spherical 40, 200, 400 μg/kg/day, intraperitoneal injection 8 days Sodium citrate –53 mV No toxicity; normal behavior, weight, serum biochemistry, hematology, histology, despite organ accumulation. Dose-dependent bioaccumulation occurred, but no adverse effects; supports therapeutic applications.

22. Toxicity in Zebrafish.

Sr. No. organism particle size & shape method of synthesis dosage & route exposure duration capping/reducing agent ζ-potential observed toxic effect outcome references
1 Zebrafish embryos Citrate/TPPMS AuNPs; 3–100 nm; spherical Citrate reduction, tannic acid 0.25–250 μM; waterborne 4–120 hpf Uncapped/TPPMS   <3% mortality, no morphological effects Inert; low toxicity
2 Adult zebrafish Biogenic AuNPs (Acalypha indica); <30 nm; spherical Leaf extract (green synthesis) 9.7–58.2 mg/L; waterborne 96 h LC50: 41 mg/L; 14 days Plant phytochemicals   Minimal gill/liver changes; no ROS or genotoxicity High biocompatibility
3 Zebrafish embryos TMAT-AuNPs; 1.3 nm; spherical Ligand exchange 0.08–50 mg/L; injection   TMAT Positive Eye/behavioral defects, apoptosis, axonal inhibition, gene suppression Surface charge key to toxicity
4 Zebrafish embryos BRP-AuNPs; 6.34 ± 2.01 nm; spherical Green synthesis with Brazilian red propolis Dose-dependent; waterborne   Natural BRP ligands –17.2 mV Oxidative stress, developmental delays, morphology anomalies Green synthesis reduces but not eliminate toxicity
5 Zebrafish embryos, ZF4 GO–Au nanohybrids; AuNPs embedded in GO AuNPs into graphene oxide ≤150 μg/mL; cell/embryo exposure   GO–Au hybrid   Lower ROS/apoptosis than GO alone; improved dispersion Safer nanohybrid alternative
6 Zebrafish embryos Au@CT; 7.6 ± 2.2 nm; spherical Marine algae extract (green synthesis) 1.25–2.5 μM; waterborne   Plant-based –24.6 mV Low embryotoxicity; minor heart rate/epiboly effects Safer vs raw extract/Au@CB
7 Zebrafish embryos Spinacia oleracea AuNPs; 20–30 nm; spherical Leaf extract; HR-TEM, UV–Vis Up to 300 μg/mL; waterborne   Plant phytochemicals Likely reduced 100% mortality at high dose; moderate anomalies; better than AgNPs Moderate toxicity; better than silver
8 Transgenic zebrafish Functionalized AuNPs; 10–100 nm; spherical PEG, TNFa, NHS/PAMAM IV injection   PEG, TNFa, PAMAM Variable Kidney/gut uptake; no oxidative or renal toxicity High uptake; low acute toxicity
9 Zebrafish embryos Citrate AuNPs; 11.6 nm; spherical Citrate reduction Up to 1.2 nM; passive diffusion via chorion 120 hpf None reported   24% mortality; 2% deformities; 74% normal Random accumulation affects outcomes
10 Zebrafish (larvae/adults) Casein-coated AuNPs; ∼5 nm; spherical NaBH4 reduction with casein Intracardial injection   Casein protein Neutralized Crossed BBB; no toxicity; reversed Aβ-induced damage Potential for neurotherapy
11 Zebrafish embryos Large AuNPs; 86.2 ± 10.8 nm; spherical Citrate reduction 0–20 pM; aquatic incubation   Not reported Implied stable >96% developed normally; minimal deformation Large particles may be more biocompatible
12 Zebrafish embryos Ammannia baccifera AuNPs; size not reported; spherical Green synthesis Dose-dependent; waterborne   Plant phytochemicals –25.801 mV Accumulation; immune gene activation; moderate mortality Dose threshold important
13 Zebrafish embryos Drug-loaded AuNPs; 20–30 nm; spherical Sunlight-mediated with 6-MP, folic acid Immersion   Nutrient agar + drugs Likely reduced Minimal toxicity; low stress from fluorescence and heartbeat Surface function lowers embryonic stress
14 Zebrafish embryos Pectin-mediated AuNPs; 4–18 nm; polymorphic/polydisperse Green synthesis using pectin Dose-dependent; waterborne   Pectin –33.4 mV Higher doses increased mortality; minimal genotoxicity at low doses Important to control concentration

23. In Vitro Toxicity of AuNPs Tested on Various Cell Lines.

Sr. No. organism particle size & shape method of synthesis dosage & route exposure duration capping/reducing agent ζ-potential observed toxic effect outcome references
1. BEAS-2B, HEK 293, CHO AuNPs (14 and 20 nm, spherical) Citrate reduction In vitro, various doses 24 h Citrate –30 mV (typical) Mild cytotoxicity in BEAS-2B/HEK 293, higher in CHO Uptake rapid; size/toxicity correlation weak
2. A549, NCIH441 AuNPs (27–28 nm, spherical) Tetrachloroauric acid + sodium citrate 0.7 mM, in vitro Up to 72 h Sodium citrate –13 mV (before) to –12 mV (after) Viability >60%; A549 more sensitive Surface chemistry affects toxicity
3 DU145, MDA-MB-231, L132 Aurovist (1.9 nm, spherical) Commercial Up to 13,300 μg/mL 24 h Not specified Highly negative (typical) Tumor-specific apoptosis, ROS generation High selectivity, dose- and cell-specific toxicity
4 Vero, NIH3T3, PK-15, MRC-5 Commercial AuNPs (∼10–40 nm, nanorod) Commercial         Apoptosis/autophagy/cell cycle arrest Cell-line specific stress responses.
5 HT29 AuNPs (<100 nm, spherical) Not specified 2–10 μg/mL     Measured, not stated Moderate cytotoxicity, no DNA damage Oral route implications suggested
6 MCF-7, HepG2, HCT-116 AuNPs (3.4–6.9 nm, spherical/triangular) Green synthesis using flaxseed extract In vitro, various doses   Lignans (flaxseed) Implied stable Dose-dependent tumor cytotoxicity Effective anticancer potential
7 MRC-5, Mice AuNPs (15–20 nm, spherical) Chemical synthesis In vitro + oral (mice)   Aspartic acid, citrate, BSA Varied; but not given in numeric GNPA/GNPC mild liver/kidney toxicity; GNPB nontoxic Capping agent crucial for stability
8 DU-145 AuNPs (20 and 200 nm, spherical) Commercial In vitro, high doses 48–72 h Mannose-functionalized and uncoated   20 nm more toxic; enhanced uptake Size & surface mod. affect diagnostics
9 MCF-7 AuNPs (∼20 nm, spherical) Citrate reduction + alginate coating 12 nM, in vitro   Sodium alginate Stable postcoating Viability ∼29%; oxidative stress pH-responsive release mechanism
10 Hepatocellular carcinoma (C3A) AuNPs (40 and 80 nm, spherical) Chemical Not specified   BPEI, LA, PEG Charge dependent PEG-AuNPs biocompatible; BPEI toxic Corona formation modulates toxicity
11 MCF-7, MDA-MB-231 AuNPs (∼narrow, spherical) Green synthesis via *Curcuma caesia* In vitro, low μg/mL Not specified Phytochemicals Implied stable Selective toxicity to cancer cells Apoptosis-mediated effect
12 3T3 Fibroblasts AuNPs (∼9 nm, spherical) Chemical In vitro   Glutathione Implied stable Minimal cytotoxicity; low ROS Dual antioxidant and nanocarrier role
13 HepG2 AuNPs (118 ± 72 nm, irregular) Green synthesis (*Verbascum speciosum*) In vitro, various doses Time/dose dependent Phytochemicals Not mentioned Antiproliferative effects Likely apoptosis/ROS-based
14 Not specific AuNPs (<100 nm, various) General/Review Environmental Chronic/unknown Various Unstable small ones risky Oxidative stress, inflammation Smaller poorly capped particles most toxic
15 HepG2, MCF7, HeLa, Vero AuNPs (25–35 nm, spherical) Green synthesis using *Cassia roxburghii* Up to 75 μg/mL In vitro Phytochemicals Not disclosed Selective toxicity (IC50 30–50 μg/mL) DNA fragmentation; apoptotic mechanism
16 THP-1 AuNPs (<50 nm, spherical) Green synthesis using *Centaurea behen* In vitro   Phytochemicals   IC50 ∼ 25 μg/mL Mild antioxidant, strong anticancer
17 DU-145, PC-3, MDA-MB-231 GNR & GNS (shape-specific) Chemical (CTAB & citrate) 0.55–34.5 μg/mL In vitro BSA Surface coating dependent GNR > GNS cytotoxicity; shape-specific GNR reduced migration; therapeutic potential
18 8 Mammalian Cell Lines Au-PEI (∼10–20 nm, cationic) & Au-PEG-COOH (anionic) Chemical Various doses In vitro PEI, PEG Positive (PEI), Negative (PEG) PEI cytotoxic; PEG biocompatible Model systems for uptake/toxicity
18 H69 AuNPs (<40 nm, spherical) Green (*Coffea arabica*), drug-loaded In vitro, Dox-conjugated   Phytochemicals + MPSNa/MUA Stable Enhanced cytotoxicity vs free Dox pH-dependent release; lower off-target toxicity
19 HepG2 AuNPs (10–50 nm, polydispersed) Biosynthesis via *Enterococcus sp.* In vitro, 5–15 μg/mL   Bacterial enzymes - Mitochondrial apoptosis; ROS, cytochrome c Effective green therapy
20 HepaRG, Primary Rat Hepatocytes AuNPs; 15 and 60 nm spheres/stars Turkevich & Bastus In vitro with/without FBS   Citrate, MUA Inferred via stability Smaller, citrate-capped AuNPs more toxic; MUA-coated safer Serum impacts toxicity and uptake

24. Toxicity of AuNPs with Organism, Cell Type, Toxicity Effects, and Significance.

Sr. No. organism cell type particle shape toxicity and effects particle size of particular type significance references
1 Zebrafish embryos Embryonic cells Spherical Dose-dependent toxicity with significant effects above 40 mg/L; toxicity varies with coating agents and concentrations. Biogenic gold nanoparticles (BRP-GNPs): Size 6.34 ± 2.01 nm (TEM), Hydrodynamic diameter 58.79 ± 1.69 nm The need for concentration and coating agent considerations in assessing nanoparticle toxicity, especially for early stage developmental organisms.
2 Mice Phagocytic cells (liver and spleen), Renal cells Spherical Accumulation in liver, spleen, and kidneys due to slow clearance, leading to mild structural changes and increased inflammatory markers. BSA-coated gold nanoparticles (BSA-AuNPs): Gold core size 18.3 ± 1.5 nm; Hydrodynamic diameter: 38 ± 5 nm in water, 48 ± 23 nm in 5% glucose solution Emphasizes the impact of slow nanoparticle clearance and accumulation in organs, raising concerns for long-term exposure and inflammatory responses.
3 Drosophila melanogaster Hemocytes Spherical Citrate-capped AuNPs (15 nm) cause genotoxicity, leading to DNA damage and hereditary mutations. Citrate-capped gold nanoparticles (Size: 15 nm) It shows the potential genotoxic effects of nanoparticles, underlining the need for careful evaluation of their environmental and biological impact.
4 Wistar rats Hepatocytes, Renal cells, Reproductive cells Spherical Accumulate in liver, kidneys, and reproductive organs, causing oxidative stress, inflammation, and organ damage. Gold nanoparticles with a size of 5–50 nm The toxic potential of nanoparticle accumulation in organs, with implications for long-term exposure risks in mammals.
5 DU-145, PC-3 (Prostate), MDA-MB-231 (Breast) Cancer cells (Prostate, Breast) Spherical GNR-BSA had higher uptake and inhibited migration; GNS-BSA had minimal impact. GNR-BSA (127.95 nm), GNS-BSA (35 nm) The toxic potential of nanoparticle accumulation in organs, with implications for long-term exposure risks in mammals
6 Vero cells (African green monkey kidney cells) kidney cells Spherical AuNPs antibacterial activity against drug-resistant bacteria. Biosynthesized gold nanoparticles (Size: 7.1–40.0 nm, average 23.2 ± 10.7 nm) The potential of AuNPs in combating bacterial resistance, with varying effects based on particle size.
7 RAW 264.7, HaCaT, NHDF, Mice Macrophages, Keratinocytes, Fibroblasts Spherical Bifi-CKAuNPs showed no significant cytotoxicity, inhibited NF-κB/MAPK signaling, and reduced inflammation in mice lung, liver, and kidneys. Bifi-CKAuNPs size is 10–25 nm Anti-inflammatory applications with minimal cytotoxicity, highlighting the role of nanoparticles in modulating immune signaling pathways.
8 Male albino rats Hepatocytes, Renal cells, Glial cells Spherical Accumulated in liver, kidney, and brain, with higher toxicity at 100 mg/kg, causing hepatic and renal changes. 10 mg/kg showed minimal toxicity. Gold nanoparticles (Size: 7.8 nm) Highlights the toxicity of high nanoparticle concentrations, stressing the need for dose optimization in therapeutic applications.
9 BEAS-2B, CHO, HEK 293 Human embryonic kidney cells Spherical Showed minimal toxicity, with CHO cells most sensitive to 20 nm particles. Uptake varied by cell type and time. Citrate-stabilized gold nanoparticles (Size: 14 nm, 20 nm) Emphasizes the effect of particle size and cell type on nanoparticle toxicity, suggesting the importance of size and cellular context in biomedical applications.
10 Rat ovarian granulosa cells Ovarian granulosa cells Spherical Accumulated in mitochondria, lipid droplets, and vacuoles, causing structural changes. Short-term exposure increased estrogen secretion, but levels dropped after 24 h due to mitochondrial damage. 10 nm AuNPs in concentration 2.85 × 10 10/mL Shows endocrine disruption from nanoparticles, especially in reproductive health, emphasizing the need for safety evaluations in reproductive biology.
11 DLA, EAC, Swiss albino mice Normal peritoneal cells Spherical AuNPs showed no significant toxicity at 100 mM, with no metabolic changes, organ damage, or tissue abnormalities in mice. Gold nanoparticles (Size: 4.5 nm) Highlights the relatively low toxicity of small AuNPs at specific concentrations, suggesting potential biocompatibility for therapeutic uses.
12 Human (Homo sapiens) HEK 293 cells Spherical Phosphine-stabilized AuNPs blocked hERG channels, indicating cardiotoxicity; Thiol-stabilized AuNPs had no effect. Gold nanoparticles (1.4 nm), phosphine-stabilized and thiol-stabilized Highlights the significance of surface stabilization in nanoparticle toxicity, particularly in relation to cardiovascular health.
13 Human (Homo sapiens) and Mice MRC-5 human fibroblast cells Spherical No significant cytotoxicity in MRC-5 cells; GNPC and GNPA induced mild hepatotoxicity and nephrotoxicity in mice; GNPB was the most biocompatible; Stability varied with capping agents, influencing biomedical potential Gold nanoparticles (15–20 nm), aspartic acid-capped (GNPA), trisodium citrate-capped (GNPC), and bovine serum albumin-capped (GNPB) Emphasizes the importance of nanoparticle surface chemistry in biocompatibility and potential biomedical applications, such as drug delivery and therapy.
14 Human (Homo sapiens) HT29 human colorectal adenocarcinoma cells Spherical Intracellular accumulation but did not induce significant DNA damage or apoptosis; silver and metal oxide nanoparticles caused higher cytotoxicity Gold nanoparticles (32 nm) Suggests that AuNPs may have lower toxicity Compared to other metal nanoparticles, emphasizing the importance of particle size in toxicity assessments.
15 Mouse Balb/3T3 fibroblast cells Spherical AuNPs showed cytotoxicity at ≥50 μM after 72 h, disrupted actin cytoskeleton, caused cell contraction and morphology changes; 15 nm AuNPs were nontoxic; Both sizes were internalized via endocytosis, accumulating in endosomal compartments, with clathrin-mediated uptake and clathrin degradation observed Citrate-stabilized AuNPs of 5 and 15 nm diameter in concentrations of 2, 10, 20, 39.2, 58.8 g/mL Highlights size-dependent toxicity and cellular uptake mechanisms, stressing the need for further research on nanoparticle-cell interactions
16 Human (Homo sapiens) HepG2 and PBMCs cells Spherical PAMAM dendrimers induced ROS production, DNA damage, and reduced cell viability; HepG2 cells were more sensitive than PBMCs, suggesting potential for cancer therapy with minimal effects on healthy cells Gold nanoparticles (7–20 nm), citrate-coated and PAMAM dendrimer-coated Demonstrates the role of surface chemistry in nanoparticle toxicity, suggesting cancer therapy applications with minimal toxicity to healthy cells.
17 Mammalian (CHO K1, A549, HeLa, NIH 3T3, HaCaT, SK-MEL-28, BEAS-2B, HepG2) Various cell lines Spherical low toxicity up to 50 μg/mL; Toxicity was dose-dependent and linked to surface charge, with positively charged Au-PEI nanoparticles exhibiting higher cytotoxicity Gold nanoparticles (11.5–14.3 nm), PEI- and PEG-COOH-functionalized Emphasizes the role of surface charge and functionalization in modulating nanoparticle toxicity and their potential in targeted therapies.
18 Human (Homo sapiens) Hela cells Spherical No significant cytotoxicity at high concentrations; glucose-functionalized nanoparticles showed higher uptake than PEG-coated ones Gold-coated iron oxide nanoparticles (4.6–5.4 nm), PEG- and glucose-functionalized Highlights the importance of surface functionalization for improving nanoparticle uptake and biocompatibility in biomedical applications
19 Human cells LNCaP prostate cancer and HFF healthy fibroblast cells Spherical Concentration-dependent cytotoxicity; reduced cancer cell viability, but also affected healthy cells at higher doses; Protein corona reduced cytotoxicity on cancer cells Gold nanoparticles (60–250 nm) Highlights the importance of nanoparticle size, surface Interactions, and biological environment in toxicity, suggesting a need for tailored nanomedicine approaches in cancer therapy
20 Human cells HaCaT human keratinocyte cells Spherical Positively and negatively charged AuNPs caused mitochondrial stress, apoptosis, and DNA damage; neutral AuNPs induced necrosis; Cytotoxicity was dose-dependent The average particle sizes are reported as follows: 1.8 ± 0.7 nm for neutral particles (MEEE), 1.6 ± 0.8 nm for positive particles (TMAT), and 1.8 ± 0.7 nm for negative particles (MES). Highlights the role of surface charge in nanoparticle-induced cytotoxicity, emphasizing the need for safety evaluations in biomedical applications.
21 Mytilus edulis (Blue Mussels) Digestive gland cells Spherical It caused lipid peroxidation, decreased thiol-containing proteins, and reduced lysosomal membrane stability, indicating oxidative stress. Gold nanoparticles (5.3 ± 1 nm) Highlights the stronger oxidative stress induced by smaller AuNPs, emphasizing the need for further research on nanoparticle toxicity in marine organisms.
22 Rats Liver and spleen cells Spherical Rapidly cleared from bloodstream, accumulated mainly in liver and spleen. After 28 days, spleen atrophy and mild anemia observed, especially with CALNN-coated AuNPs. Gold nanoparticles (∼20 nm), Citrate- and pentapeptide CALNN-coated Surface chemistry influences toxicity more than biodistribution, emphasizing the need for improved nanoparticle coatings for biomedical applications.
23 Nicotiana xanthi (Tobacco) Seedling cells Spherical Smaller AuNPs (3.5 nm) entered roots and translocated to leaves, causing necrosis. Larger AuNPs (18 nm) remained on root surfaces, indicating size-dependent uptake and toxicity. Gold nanoparticles (3.5 and 18 nm) Highlights the importance of nanoparticle size in plant uptake and toxicity, emphasizing the need for further research on AuNP interactions with plants and their environmental impact.
24 Daphnia magna (Water flea) Aquatic organism Spherical Toxicity varied with surface charge; positively charged PAH–AuNPs caused the highest mortality, while Cit–AuNPs and MPA–AuNPs were less toxic. Chronic exposure affected reproduction, indicating long-term impacts. Gold nanoparticles (4–5 nm), PAH-, Cit-, and MPA-coated Highlights the importance of surface chemistry and charge in nanoparticle toxicity, emphasizing the need for environmental risk assessment in aquatic ecosystems.
25 BALB/c mice Various organs (Lungs, Kidneys, Liver, Spleen) Spherical No significant toxicity, immune response, or renal stress even at 60 μM concentrations. Rapid excretion through kidneys; initial accumulation in lungs and kidneys, later shifting to liver and spleen. Glutathione-coated gold nanoparticles (1.2 ± 0.9 nm) supports the use of glutathione as a biocompatible alternative to PEG for nanomedicine and targeted drug delivery.
26 Mice Macrophages Core–shell Rapidly cleared by macrophages; mild inflammation and oxidative stress, resolved in 2 weeks. No major toxicity in biochemical or histological analyses. Rectal administration showed no systemic absorption or toxicity. Raman-active silica-gold nanoparticles (60 nm gold core, 30 nm silica shell, total ∼120 nm) Suggests that while intravenous exposure may cause temporary immune activation, localized administration could be safer for biomedical applications.
27 Human Cells Lymphocytes, Fibroblasts Spherical Higher micronuclei formation in lymphocytes, slight DNA damage in fibroblasts; size- and concentration-dependent toxicity Citrate-capped gold nanoparticles (AuNPs) with sizes of 15 and 47 nm Greater genotoxicity in smaller 15 nm particles; critical for assessing biomedical applications of gold nanoparticles
28 3T3 Cells Fibroblast Cells Spherical GSH-AuNPs significantly reduced oxidative stress compared to glutathione alone and nonstabilized AuNPs Glutathione-stabilized gold nanoparticles (GSH-AuNPs) with an average size of 9 nm Potential for reducing oxidative stress in ROS-related conditions, promising for biomedical applications

Many studies have been carried out to replace these nanoparticles with biobased materials to address toxicity-related issues in various applications.

4.1. Toxicity of AuNPs on Human Health

Gold nanoparticles (AuNPs) exhibit unique biomedical potential, yet their toxicity remains a concern, as size, shape, surface chemistry, and concentration significantly influence cellular uptake, oxidative stress, and cytotoxic responses. Table depicts various factors affecting toxicity of AuNPs. Gold nanoparticles (30, 50, 90 nm) exhibited dose- and time-dependent cytotoxicity in HL-60 and HepG2 cells. Cytotoxicity correlated with oxidative stress markers, including increased ROS, GSH depletion, and altered SOD (Superoxide Dismutase) activity. NAC conferred partial protection, particularly with 30 nm particles. Effects varied slightly with nanoparticle size and cell type. Gold nanoparticles (∼20 nm) with citrate or 11-MUA coatings were tested on HepG2 cells. Both showed no cytotoxicity, but citrate-AuNPs induced DNA damage at low concentrations. Internalization was similar for both, highlighting surface coating’s role in AuNPs’ biocompatibility and genotoxic potential despite negligible cytotoxic effects. Surface charge significantly influenced the cytotoxicity of 1.5 nm gold nanoparticles in human keratinocyte (HaCaT) cells. Charged AuNPs induced dose-dependent toxicity, mitochondrial dysfunction, altered gene expression, and apoptosis, while neutral AuNPs caused necrosis, highlighting surface charge as a key factor in AuNP–cell interactions. In HL7702 human liver cells, 8 nm gold nanoparticles (AuNPs) triggered early cytosolic glutathione (GSH) depletion, leading to mitochondrial depolarization and apoptosis. Strong Au–S interactions initiated Bax translocation, H2O2 buildup, and caspase-3 activation, confirming GSH loss as a key apoptotic trigger via the mitochondrial pathway. Gold nanoparticles (AuNPs) exhibited dose-dependent cytotoxic effects on human spermatozoa, significantly reducing motility and viability at higher concentrations. AuNPs were internalized by sperm cells, indicating potential reproductive toxicity and the need for further investigation into their genotoxic effects on germ cells. AuNPs induced oxidative stress and autophagy in MRC-5 human lung fibroblasts. Increased lipid peroxidation, autophagosome formation, and upregulation of MAP-LC3, ATG7, and stress response proteins suggest AuNPs trigger oxidative damage, with autophagy acting as a protective cellular response mechanism.

4.2. Toxicity of AuNPs on Environment

Gold nanoparticles (AuNPs) are often considered less toxic than silver counterparts. Their toxicity is primarily linked to oxidative stress, disruption of cell membranes, and DNA damage. They tend to accumulate in digestive tissues, causing lipid peroxidation and disrupting in antioxidant enzyme functions. At moderate concentrations AuNP depicts low cytotoxicity, increased reactive oxygen species shows long-term threats to the environment. They depict significant environmental toxicity due to their high mobility, reactivity, and accumulation of living organisms. In aquatic systems, they are absorbed by plankton and fish, disrupting vital functions and moving through the food web. In soil, AuNPs impact plant growth and disrupts the microbial activities, altering nutrient availability. Their persistence and capacity to penetrate biological barriers that may lead to oxidative stress and DNA damage in unintended organisms. Proper regulation and eco-safe design are essential to reduce the environmental risks. Gold nanoparticles (AuNPs) are used in various industries and can enter the environment via waste streams or incorrect disposal methods. Due to their small size, they can be easily absorbed by organisms, potentially disrupting biological activities. In aquatic environments, they threaten fish and microbes by causing oxidative stress and bioaccumulation. The lack of comprehensive data on their long-term effects raises concerns, stressing the need for monitoring, product labeling, and implementing proper nanowaste management regulations to limit the environmental damage. Another study shows variable toxicity in aquatic environments, based on factors like size, shape, and surface coating. Functionalized AuNPs developed for medical applications may pose ecological hazards. In studies with microalgae and Daphnia magna, AuNPs induced oxidative stress and DNA damage. Aggregation in water does not necessarily reduce toxicity, they can move through food webs. Gold nanoparticles synthesized using Carica papaya leaf extract demonstrated an eco-friendly and effective approach for photocatalytic degradation of methylene blue, achieving 95% reduction in 90 min and 99% removal of total organic carbon. Characterization confirmed their crystalline structure, morphological, and surface characteristics.

5. Regulatory Frameworks Governing Gold Nanomaterials

Global regulations for nanomaterials differ significantly across regions. Kus-Liśkiewicz et al., in their comprehensive review, have effectively detailed the global regulatory landscape surrounding nanomaterials (Table ). They highlight the contrasting approaches across regions, noting how stringent guidelines by agencies like the FDA and EMA differ from the more lenient or absent regulations in parts of Asia and South America. The U.S. FDA and Europe’s EMA mandate stringent nanosafety evaluations for nanomaterials, particularly in food and pharmaceuticals, requiring comprehensive physicochemical, toxicological, and ecotoxicological data. In contrast, countries like China, Japan, South Korea, and Brazil often lack specific regulatory frameworks for nanomaterials in cosmetics and food. The U.S. launched the National Nanotechnology Initiative (NNI) in the year 2000 to promote nanoscience research and define “nanopharmaceuticals”. Despite ongoing advancements, major challenges remain in the classification of novel nanomaterials and the development of standardized testing methods essential for ensuring their safe commercial and clinical application. , Regulations for gold nanoparticles are gradually evolving worldwide, aiming to ensure their safe use in medical, food, and consumer products. Standardized protocols and international harmonization are crucial for effective risk assessment and commercialization.

25. Regulating Authorities That Have Defined Specific Requirements for Various Groups of Nanoproducts .

Authority Name regulations, procedures, standardisation, and references
Joint Research Centre (JRC) Provides evidence-based scientific support in EU policymaking process.
European Chemicals Agency (ECHA) Implements REACH: Registration, Evaluation, Authorisation, and Restriction of Chemicals.
Scientific Committee on Emerging and Newly Identified Health Risks (SCENIHR) Gives opinions on emerging or complex health/environmental risks needing multidisciplinary risk assessment.
Scientific Committee on Consumer Safety (SCCS) Advises on risks of nonfood consumer products like cosmetics, toys, textiles, etc.
European Food Safety Authority (EFSA) Publishes nanosafety guidelines for food products, recommends analytical technologies.
European Medicines Agency (EMA) Ensures scientific evaluation and supervision of medicines for human and animal health in the EU.
U.S. Food and Drug Administration (FDA) Protects public health through safety and efficacy of drugs, food, medical devices, and cosmetics.
Quality Supervision, Inspection, and Quarantine (AQSIQ) Oversees commodity inspection, food safety, certification, accreditation, and standardization in China Available online: https://www.aqsiq.net
Chinese National Nanotechnology Standardization Technical Committee (NSTC) Reviews nanomaterial standards. Available online: http://english.nanoctr.cas.cn/
Standardization Administration of China (SAC) Establishes standards for nanomaterials and their characterization. https://www.sac.gov.cn/
Brazilian Health Surveillance Agency (ANVISA) Regulates research, production, disposal, and use of nanotechnologies. https://www.gov.br/anvisa/pt-br
a

Adapted and modified from ref . Copyright 2021, with permission of MDPI.

6. Limitations and Future Prospects

Even though AuNPs are considered generally safe and biocompatible and also act as promising agents in biomedical fields due to their unique physicochemical properties, factors like shape, size, drug dosage, electrical charge, and purity of the gold formation can play a role in determining the cytotoxicity. The toxicity can depend on the surface chemistry of the nanoparticles, including the type and concentration of capping ligands, which can also influence their interactions with biological systems. The nanoparticles can be penetrative in nature resulting in increased threat to humans, flora and fauna, and the environment. The synthesis methods should incorporate biobased mechanisms using biobased extracts, hence minimizing the toxic effects. Also, surface modifications to the AuNPs and aggregations significantly affect their functionality and reduce their effectiveness. New researchers should aim to find advanced and green methods of synthesis that aim to improve the health and drug delivery effectiveness, improve sensing applications, incorporate models for safe environmentally friendly catalytic applications, and reduce toxicity. , AuNPs have a high potential application in next-generation electrical sensors, biomedical devices, solar energy harvesting, precision gene therapy delivery methods, advanced anticancer diagnostics, and health and sanitation applications. Future research should focus on optimizing the synthesis strategies of gold nanoparticles (AuNPs) to enhance their stability, biocompatibility, and functionality for diverse biomedical applications. Comparative studies, such as those evaluating sonochemical and chemical reduction methods, have demonstrated that synthesis techniques directly influence the physicochemical characteristics and antibacterial activity of AuNPs. Building on this, integrating multifunctional nanostructures such as Fe3O4@Au hybrids holds promise for advanced applications in drug delivery, catalysis, hyperthermia, and imaging. , Recent advancements in gold nanoparticle (AuNP) research highlight green mycosynthesis using mushrooms like Agaricus bisporus and rapid sonochemical techniques as promising routes for eco-friendly and scalable production. These methods yield stable, crystalline AuNPs with diverse morphologies and excellent catalytic and biomedical properties, including CT contrast enhancement and methylene blue degradation. Core@shell Fe3O4@Au structures synthesized via optimized sonochemical methods show dual-mode MRI/CT imaging potential. Sonochemistry also outperforms laser ablation in purity and colloidal stability. Furthermore, the development of eco-friendly, green synthesis methods using biological extractssuch as plant or mushroom derivativesnot only reduces environmental toxicity but also improves the therapeutic potential of AuNPs in anticancer treatments. Gold nanoparticles (AuNPs) synthesized using fresh fruiting bodies of Enoki and Shiitake mushrooms as green reducing agents exhibited diverse shapes, good stability, and crystallinity, with sizes around 72–74 nm. These mycosynthesized AuNPs showed effective catalytic degradation of Methylene Blue and potential for biomedical applications. Sonochemical methods further enhanced nanoparticle dispersion and enabled Au coating on Fe3O4 via acoustic cavitation. Ultrasmall AuNPs derived from fruit peels, for example, demonstrate strong photothermal effects and enhanced biocompatibility, offering a sustainable pathway for developing next-generation nanomedicine.

6.1. Emerging Trends

Nowadays, Two-dimensional (2D) gold (Au) nanocrystals have been an area of interest for researchers. Gold nanoparticles are also used in antiaging creams or masks having particle size 5–400 nm, which enhances the collagen formation by 20–200 times. Cosmetic companies such as L’Oreal and L’Core Paris have infused gold nanoparticles in their formulations making them more effective. Recent advancements in biosensor and bioelectronics technology have opened new possibilities in biomedical diagnostics, particularly for early cancer detection. Biosensors are being used for detection of cancer with the use of advance techniques. Fox et al. has done a detailed investigation into the optimized synthesis and mechanism of formation for ultrathin, free-standing two-dimensional gold nanosheets (AuNS), which are approximately 0.47 nm thick and shows aspiring results for applications in computing, biosensing, catalysis, and healthcare. The AuNS were synthesized by one-pot, seedless reaction using chloroauric acid (HAuCl4) reduced by sodium citrate in the presence of methyl orange (MO). Spectrophotometric and TEM analyses confirmed that MO is rapidly oxidized to form 4-diazobenzenesulfonic acid (4-DBSA), and this oxidation is crucial to directing two-dimensional growth. However, by using 4-DBSA alone can yield low-quality AuNS, showing that MO’s self-assembly and in situ oxidation synergistically assists nanosheet formation. UV–vis and other kinetic studies showed the MO degradation during the process follows pseudo-first-order kinetics and occurs within 30 s. Optimization such as delaying the addition of reagents by 30 s and slow mixing were observed for higher yields. Also, by lowering the reaction temperature from 20 °C to 4 °C can enhance AuNS yield by 16 times. The synthesis was complete within 8 h, with nanosheets observable after just 1 h. Mild centrifugation (∼1000 g) was essential to isolate the AuNS over 3D structures. The resulting AuNS showed enhanced catalytic activity compared to spherical gold nanoparticles,which makes them highly attractive for use in catalytic, enzymatic, and diagnostic applications due to their large surface area-to-volume ratio. A study conducted by Huang et al. presents a novel yet cost-effective way to develop an electrochemical biosensor for the ultrasensitive detection of hydrogen peroxide (H2O2), a cancer biomarker, especially used in identifying colon cancer cells. The sensor utilizes a dual-nanozyme system by combination of ultrathin 2D conductive Cu-HHTP nanosheets with densely deposited ultrafine gold nanoparticles of size 3 nm. The Cu-HHTP nanosheets provide a large surface area and active Cu–O4 metal sites which enhances catalytic activity, while the embedded AuNPs prevent from aggregation and further amplifying the response. This synergistic nanohybrid structure offers exceptional electrocatalytic efficiency, achieving an exceptionally low detection limit of 5.6 nM and a high sensitivity of 188.1 μA cm–2 mM–1. This biosensor successfully tracks real-time H2O2 release in human colon cells, by distinguishing cancerous cells from normal epithelial cells. It exhibits strong selectivity, reproducibility, and stability. This is a promising approach for advancing early cancer diagnostics and could inspire further development of nanozyme-based biosensors in biomedical and clinical applications. Another approach is made by Rauf et al. to develop an electrochemical biosensor using laser-scribed graphene modified with gold nanostructures (LSG-AuNS) to address the limitations of bare LSG electrodes in sensitivity and biomolecule immobilization. Gold nanostructures were electrodeposited onto LSG, which enhances the sensitivity and catalytic activity almost by 2-fold compared to unmodified LSG and commercial screen-printed gold electrodes. This 3D-porous LSG-AuNS platform was utilized to fabricate an aptamer-based sensor for detection of the breast cancer biomarker HER2, achieving a detection limit of 0.008 ng/mL and a broad linear detection range of 0.1–200 ng/mL. This sensor effectively detected HER2 in undiluted human serum with high accuracy. Furthermore, the system was integrated into a hand-held electrochemical point-of-care (POC) device, controlled via a custom mobile application. Although minor nonspecific protein adsorption was noted, the platform demonstrated high potential toward biosensing applications. A highly sensitive and multiplex electrochemical biosensor was developed for detecting multiple cancer-related microRNAs (miRNAs) in plasma by utilizing a dual amplification strategy. This approach by Mohammadniaei et al. combines MXene-Ti3C2T x nanosheets decorated with 5 nm gold nanoparticles (AuNPs) for enhanced signal transduction and duplex-specific nuclease (DSN)-based target recycling for signal amplification. Colorimetric biosensors rely on the visible color change of gold nanoparticles (AuNPs) in response to the presence or absence of a target analyte. This method offers a simple but yet fast approach for cancer detection. AuNPs are widely used in these sensors due to their unique optical properties, allowing for rapid and easily detectable color shifts that provide a quick visual indication of the analyte. A novel and highly sensitive colorimetric biosensor was developed by Xiao et al. using gold nanoparticle-decorated Bi2Se3 (Au/Bi2Se3) nanosheets, synthesized through simple sonication of a gold precursor with Bi2Se3 in water. Bi2Se3 nanosheets exhibit excellent electron-donating properties also it synergizes with AuNPs to enhance catalytic performance, particularly in the 4-nitrophenol (4-NP) reduction by NaBH4. The sensor works through a smart “on–off” mechanism: its catalytic activity is briefly turned off when it comes into contact with certain cancer-related antibodies, such as anti-CEA. However, this activity switches back on when the matching antigen like CEA is present. This clever response allows the system to precisely detect cancer biomarkers. Impressively, it can identify carcinoembryonic antigen (CEA) at concentrations as low as 160 picograms per milliliter, demonstrating strong potential for use in real-world clinical diagnostics. The biosensor is versatile for detection of other cancer markers like AFP and PSA. The work introduces a promising platform for biosensing, with future potential in diagnostics, biocatalysis, and smart biomaterial systems, Currently, breast cancer detection methods like mammography, MRI, and biopsy are expensive, invasive, and time-consuming, often yielding inconclusive results. To address these limitations, Selwyna et al. developed a sensitive, label-free electrochemical immunosensor that detects breast cancer by targeting the CA 15–3 biomarker in serum. The sensor uses gold nanoparticles coated with CA 15–3 antibodies, which enhance conductivity and provide a larger surface area for biomolecular interaction. In this setup, nanoparticles were anchored onto a screen-printed carbon electrode (SPCE), and the detection of antigen–antibody interactions is carried out with electrochemical impedance spectroscopy (EIS) and a potentiostat. The immunosensor effectively identifies CA 15–3 levels within a concentration range of 5 to 75 U/mL. Ranjan et al. developed a highly sensitive electrochemical immunosensor for detection of the CD44 antigen, a key breast cancer biomarker. The sensor was based on a hybrid nanocomposite comprising graphene oxide (GO), an ionic liquid (BMIM.BF4), and gold nanoparticles (AuNPs), all immobilized on a glassy carbon electrode (GCE). Graphene oxide (GO) provides numerous oxygen-rich functional groups that facilitate effective antibody attachment. At the same time, the ionic liquid and gold nanoparticles (AuNPs) work together to enhance electron conductivity and expand the electrode’s surface area. These properties combination creates a synergistic effect that greatly improves the overall sensitivity and efficiency of the sensor. This sensor showed the detection range of 5.0 fg/mL to 50.0 μg/mL in both phosphate-buffered saline and human serum.

7. Conclusions

In conclusion, gold nanoparticles (AuNPs) have emerged as a highly versatile and valuable nanomaterial due to their unique physicochemical properties, such as high surface area, tunable size, and excellent biocompatibility. As a multidisciplinary subdomain, nanobiotechnology epitomizes the amalgamation of sciences and technologies to produce nano-objects with profound applications. Among nanomaterials, nanoparticles particularly gold nanoparticles (AuNPs) have taken center stage due to their remarkable properties and versatility. The fascination with AuNPs stems from their unique characteristics, which span a wide range of shapes and sizes, enabling diverse and impactful applications. The type of synthesis and reaction conditions mediate how the properties of the AuNPs formed vary. The applications of AuNPs span various fields, including healthcare, drug delivery, catalysis, textiles, agriculture, sensing, and electronics, which makes them pivotal in advancing nanotechnology. The ease of functionalization allows for targeted interactions in biological systems, while their optical properties enable sensitive detection methods. In this review, we have delved deep into the role of AuNPs in nanotechnology, from different types of synthesis routes and properties to the factors affecting them, and their versatile range of applications and toxicity. Overall, nanotechnology, particularly gold nanoparticles, continues to drive scientific research and technological innovation, paving the way for transformative applications across various disciplines. Continued research is essential to optimize their synthesis, minimize toxicity, and unlock their full potential across industries.

Acknowledgments

The author SA would like to extend his appreciation to the Deanship of Research and Graduate Studies at King Khalid University for funding this work through Large Research Project under grant number RGP2/509/46

∥.

A.S., S.M., and N.S. contributed equally to this work. Conceptualization, S.M., A.S., T.R., N.S., N.S. R.D., and S.A.; writingOriginal draft preparation, S.A. and A.P.; writingreview and editing, T.R. and A.S.; and visualization, A.P. and S.M. All authors have read and agreed to the submitted version of the manuscript.

Large Research Project under grant number RGP2/509/46.

The authors declare no competing financial interest.

References

  1. Khan, W. S. ; Asmatulu, E. ; Asmatulu, R. . Nanotechnology Emerging Trends, Markets and Concerns. In Nanotechnology Safety, 2nd ed.; Asmatulu, R. ; Khan, W. S. ; Asmatulu, E. , Eds.; Elsevier, 2025; Chapter 1, pp 1–21. [Google Scholar]
  2. Dagar, G. ; Bagchi, G. . Nanoparticles as Potential Endocrine Disruptive Chemicals. In NanoBioMedicine; Springer: Singapore, 2020; pp 411–429. [Google Scholar]
  3. Zhang X., Guo Q., Cui D.. Recent Advances in Nanotechnology Applied to Biosensors. Sensors. 2009;9:1033–1053. doi: 10.3390/s90201033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Zafar, S. ; Rana, N. . The Convergence of Nanotechnology and Artificial Intelligence: Unlocking Future Innovations Recent Innovations Chem. Eng. 2025; Vol. 18 10.2174/0124055204359215250127071256. [DOI] [Google Scholar]
  5. Jalalvand A. R., Karami M. M.. Roles of Nanotechnology in Electrochemical Sensors for Medical Diagnostic Purposes: A Review. Sens. Bio-Sens. Res. 2025;47:100733. doi: 10.1016/j.sbsr.2024.100733. [DOI] [Google Scholar]
  6. Butt A., Bach H.. Advancements in Nanotechnology for Diagnostics: A Literature Review, Part II: Advanced Techniques in Nuclear and Optical Imaging. Nanomedicine. 2025;20(2):183–206. doi: 10.1080/17435889.2024.2439778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Ghutepatil P. R., Khapare S., Joshi R. B., Maniyar K. G.. A Critical Review of Nanotechnology in Various Fields of Engineering. AIP Conf. Proc. 2024;3149(1):030002. doi: 10.1063/5.0224984. [DOI] [Google Scholar]
  8. Jana R., Das S.. A Review on Nanotechnology for Catalysis and Solar Energy Conversion. Int. J. Renewable Energy Commer. 2024;10:32–48. [Google Scholar]
  9. Pan X., Zhou H., Li Z., Huang W., Zhao Y., Huang Y., Liang J., Wu J., Huang R., Wu M., Hu R., Fang Y., Zhao W.. Application and Safety Research of Nanotechnology in Cosmetics: Current Status, Challenges, and Future Prospects. BioNanoScience. 2025;15:284. doi: 10.1007/s12668-025-01884-9. [DOI] [Google Scholar]
  10. Prasad R. D., Prasad N. R., Prasad N., Prasad R. S., Prasad S. R., Shrivastav O. P., Prasad R. B., Prasad R. A., Prasad R. G., Saxena P., Prasad R. R., Saxena U. R., Gour M., Shaikh Y. I., Ali M. S., Algadi H., Guo X.. A Review on Aspects of Nanotechnology in Environmental Science and Engineering. ES General. 2025;7:1397. doi: 10.30919/esg1397. [DOI] [Google Scholar]
  11. del Rosario Herrera-Rivera M., Torres-Arellanes S. P., Cortés-Martínez C. I., Navarro-Ibarra D. C., Hernández-Sánchez L., Solis-Pomar F., Pérez-Tijerina E., Román-Doval R.. Nanotechnology in Food Packaging Materials: Role and Application of Nanoparticles. RSC Adv. 2024;14:21832–21858. doi: 10.1039/d4ra03711a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bumbudsanpharoke N., Harnkarnsujarit N., Gilchrist J. F., Ko S., Keawpetch T.. The Rise of Nanotechnology in Pharmaceutical and Healthcare Packaging: Applications and Future Prospective. Packag. Technol. Sci. 2025;38(1):3–31. doi: 10.1002/pts.2848. [DOI] [Google Scholar]
  13. Chen H., Luo K., Xie C., Zhou L.. Nanotechnology of Carbon Dots with Their Hybrids for Biomedical Applications: A Review. Chem. Eng. J. 2024;496:153915. doi: 10.1016/j.cej.2024.153915. [DOI] [Google Scholar]
  14. Awlqadr F. H., Majeed K. R., Altemimi A. B., Hassan A. M., Qadir S. A., Saeed M. N., Faraj A. M., Salih T. H., Abd Al-Manhel A. J., Najm M. A. A., Tsakali E., Van Impe J. F. M., Abd El-Maksoud A. A., Abedelmaksoud T. G.. Nanotechnology-Based Herbal Medicine: Preparation, Synthesis, and Applications in Food and Medicine. J. Agric. Food Res. 2025;19:101661. doi: 10.1016/j.jafr.2025.101661. [DOI] [Google Scholar]
  15. Sati A., Ranade T. N., Mali S. N., Yasin H. K. A., Samdani N., Satpute N. N., Yadav S., Pratap A. P.. Silver Nanoparticles (AgNPs) as Potential Antiviral Agents: Synthesis, Biophysical Properties, Safety, Challenges and Future DirectionsUpdate Review. Molecules. 2025;30:2004. doi: 10.3390/molecules30092004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Sati, A. ; Mali, S. N. ; Ranade, T. N. ; Yadav, S. ; Pratap, A. . Silver Nanoparticles (AgNPs) as a Double-Edged Sword: Synthesis, Factors Affecting, Mechanisms of Toxicity and Anticancer PotentialsAn Updated Review till March 2025 Biol. Trace Elem. Res. 2025. 10.1007/s12011-025-04688-w. [DOI] [PubMed]
  17. Subhan M., Choudhury K., Neogi N.. Advances with Molecular Nanomaterials in Industrial Manufacturing Applications. Nanomanufacturing. 2021;1(2):75–97. doi: 10.3390/nanomanufacturing1020008. [DOI] [Google Scholar]
  18. Malik S., Muhammad K., Waheed Y.. Nanotechnology: A Revolution in Modern Industry. Molecules. 2023;28:661. doi: 10.3390/molecules28020661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Rambaran T., Schirhagl R.. Nanotechnology from Lab to Industry - a Look at Current Trends. Nanoscale Adv. 2022;4:3664–3675. doi: 10.1039/d2na00439a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Sati A., Ranade T. N., Mali S. N., Ahmad Yasin H. K., Pratap A.. Silver Nanoparticles (AgNPs): Comprehensive Insights into Bio/Synthesis, Key Influencing Factors, Multifaceted Applications, and ToxicityA 2024 Update. ACS Omega. 2025;10(8):7549–7582. doi: 10.1021/acsomega.4c11045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Das U., Mali S. N., Mandal S.. Unveiling the Ameliorative Effects of Phyto-Selenium Nanoparticles (PSeNPs) on Anti-Hyperglycemic Activity and Hyperglycemia Irradiated Complications. BioNanoScience. 2025;15:230. doi: 10.1007/s12668-025-01830-9. [DOI] [Google Scholar]
  22. Bai X., Wang Y., Song Z., Feng Y., Chen Y., Zhang D., Feng L.. The Basic Properties of Gold Nanoparticles and Their Applications in Tumor Diagnosis and Treatment. Int. J. Mol. Sci. 2020;21:2480. doi: 10.3390/ijms21072480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Kesharwani P., Ma R., Sang L., Fatima M., Sheikh A., Abourehab M. A. S., Gupta N., Chen Z. S., Zhou Y.. Gold Nanoparticles and Gold Nanorods in the Landscape of Cancer Therapy. Mol. Cancer. 2023;22:98. doi: 10.1186/s12943-023-01798-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Georgeous J., AlSawaftah N., Abuwatfa W. H., Husseini G. A.. Review of Gold Nanoparticles: Synthesis, Properties, Shapes, Cellular Uptake, Targeting, Release Mechanisms and Applications in Drug Delivery and Therapy. Pharmaceutics. 2024;16:1332. doi: 10.3390/pharmaceutics16101332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Amendola V., Pilot R., Frasconi M., Maragò O. M., Iatì M. A.. Surface Plasmon Resonance in Gold Nanoparticles: A Review. J. Phys.: Condens. Matter. 2017;29:203002. doi: 10.1088/1361-648X/aa60f3. [DOI] [PubMed] [Google Scholar]
  26. Das M., Shim K. H., An S. S. A., Yi D. K.. Review on Gold Nanoparticles and Their Applications. Toxicol. Environm. Health Sci. 2011;3:193–205. doi: 10.1007/s13530-011-0109-y. [DOI] [Google Scholar]
  27. Elahi N., Kamali M., Baghersad M. H.. Recent Biomedical Applications of Gold Nanoparticles: A Review. Talanta. 2018;184:537–556. doi: 10.1016/j.talanta.2018.02.088. [DOI] [PubMed] [Google Scholar]
  28. Dykman L., Khlebtsov N.. Gold Nanoparticles in Biomedical Applications: Recent Advances and Perspectives. Chem. Soc. Rev. 2012;41(6):2256–2282. doi: 10.1039/C1CS15166E. [DOI] [PubMed] [Google Scholar]
  29. Yesildag C., Ouyang Z., Zhang Z., Lensen M. C.. Micro-Patterning of PEG-Based Hydrogels with Gold Nanoparticles Using a Reactive Micro-Contact-Printing Approach. Front. Chem. 2019;6:667. doi: 10.3389/fchem.2018.00667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Ealias A. M., Saravanakumar M. P.. A Review on the Classification, Characterisation, Synthesis of Nanoparticles and Their Application. IOP Conf. Ser.: Mater. Sci. Eng. 2017;263:032019. doi: 10.1088/1757-899X/263/3/032019. [DOI] [Google Scholar]
  31. Nguyen M. T., Yonezawa T.. Sputtering onto a Liquid: Interesting Physical Preparation Method for Multi-Metallic Nanoparticles. Sci. Technol. Adv. Mater. 2018;19(1):883–898. doi: 10.1080/14686996.2018.1542926. [DOI] [Google Scholar]
  32. Ealia S. A. M., Saravanakumar M. P.. A Review on the Classification, Characterisation, Synthesis of Nanoparticles and Their Application. IOP Conf. Ser. Mater. Sci. Eng. 2017;263(3):032019. doi: 10.1088/1757-899X/263/3/032019. [DOI] [Google Scholar]
  33. Odularu A. T.. Metal Nanoparticles: Thermal Decomposition, Biomedicinal Applications to Cancer Treatment, and Future Perspectives. Bioinorg. Chem. Appl. 2018;2018(1):9354708. doi: 10.1155/2018/9354708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Korshed P., Li L., Ngo D. T., Wang T.. Effect of Storage Conditions on the Long-Term Stability of Bactericidal Effects for Laser Generated Silver Nanoparticles. Nanomaterials. 2018;8(4):218. doi: 10.3390/nano8040218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Sportelli M. C., Izzi M., Volpe A., Clemente M., Picca R. A., Ancona A., Lugarà P. M., Palazzo G., Cioffi N.. The Pros and Cons of the Use of Laser Ablation Synthesis for the Production of Silver Nano-Antimicrobials. Antibiotics. 2018;7:67. doi: 10.3390/antibiotics7030067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Huang H., Lai J., Lu J., Li Z.. Pulsed Laser Ablation of Bulk Target and Particle Products in Liquid for Nanomaterial Fabrication. AIP Adv. 2019;9(1):015307. doi: 10.1063/1.5082695. [DOI] [Google Scholar]
  37. Xu C., De S., Balu A. M., Ojeda M., Luque R.. Mechanochemical Synthesis of Advanced Nanomaterials for Catalytic Applications. Chem. Commun. 2015;51(31):6698–6713. doi: 10.1039/C4CC09876E. [DOI] [PubMed] [Google Scholar]
  38. Schreyer H., Eckert R., Immohr S., de Bellis J., Felderhoff M., Schüth F.. Milling Down to Nanometers: A General Process for the Direct Dry Synthesis of Supported Metal Catalysts. Angew. Chem., Int. Ed. 2019;58(33):11262–11265. doi: 10.1002/anie.201903545. [DOI] [PubMed] [Google Scholar]
  39. Medici S., Peana M., Nurchi V. M., Lachowicz J. I., Crisponi G., Zoroddu M. A.. Noble Metals in Medicine: Latest Advances. Coord. Chem. Rev. 2015;284:329–350. doi: 10.1016/j.ccr.2014.08.002. [DOI] [Google Scholar]
  40. Sylvestre J. P., Poulin S., Kabashin A. V., Sacher E., Meunier M., Luong J. H. T.. Surface Chemistry of Gold Nanoparticles Produced by Laser Ablation in Aqueous Media. J. Phys. Chem. B. 2004;108(43):16864–16869. doi: 10.1021/jp047134. [DOI] [Google Scholar]
  41. Mafuné, Kohno F., Takeda Y., Kondow T., Sawabe H.. Formation of Gold Nanoparticles by Laser Ablation in Aqueous Solution of Surfactant. J. Phys. Chem. B. 2001;105(22):5114–5120. doi: 10.1021/jp0037091. [DOI] [Google Scholar]
  42. Resta V., Afonso C. N., Piscopiello E., Van Tendeloo G.. Role of Substrate on Nucleation and Morphology of Gold Nanoparticles Produced by Pulsed Laser Deposition. Phys. Rev. B. 2009;79(23):235409. doi: 10.1103/PhysRevB.79.235409. [DOI] [Google Scholar]
  43. Donnelly T., Krishnamurthy S., Carney K., McEvoy N., Lunney J. G.. Pulsed Laser Deposition of Nanoparticle Films of Au. Appl. Surf. Sci. 2007;254(4):1303–1306. doi: 10.1016/j.apsusc.2007.09.033. [DOI] [Google Scholar]
  44. Jankowski K., Jabłońska J., Uznański P., Całuch S., Szybowicz M., Brzozowski R., Ostafin A., Kwaśny M., Tomasik M.. Necked Gold Nanoparticles Prepared by Submerged Alternating Current Arc Discharge in Water. RSC Adv. 2022;12(52):33955–33963. doi: 10.1039/D2RA06050G. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Endla P., Radhika V.. Fabrication and Evaluation of Gold Nanoparticles by Ball Milling, Hall-Williamson and x-Ray Diffraction Method. Mater. Today: Proc. 2021;47:4993–4995. doi: 10.1016/j.matpr.2021.04.451. [DOI] [Google Scholar]
  46. Hatakeyama Y., Onishi K., Nishikawa K.. Effects of Sputtering Conditions on Formation of Gold Nanoparticles in Sputter Deposition Technique. RSC Adv. 2011;1(9):1815–1821. doi: 10.1039/c1ra00688f. [DOI] [Google Scholar]
  47. Gromov D. G., Dubkov S., Gavrilov S. A.. Features of the Nanoparticles and Binary Nanoalloys Formation during Thermal Evaporation and Condensation on an inert surface in vacuum. Technol. Process Route. 2024;45:49–58. doi: 10.24151/1561-5405-2023-28-1-49-58. [DOI] [Google Scholar]
  48. Malik M. A., Wani M. Y., Hashim M. A.. Microemulsion Method: A Novel Route to Synthesize Organic and Inorganic Nanomaterials: 1st Nano Update. Arabian J. Chem. 2012;5(4):397–417. doi: 10.1016/j.arabjc.2010.09.027. [DOI] [Google Scholar]
  49. Solanki J. N., Murthy Z. V. P.. Controlled Size Silver Nanoparticles Synthesis with Water-in-Oil Microemulsion Method: A Topical Review. Ind. Eng. Chem. Res. 2011;50(22):12311–12323. doi: 10.1021/ie201649x. [DOI] [Google Scholar]
  50. Azharuddin M., Zhu G. H., Das D., Ozgur E., Uzun L., Turner A. P. F., Patra H. K.. A Repertoire of Biomedical Applications of Noble Metal Nanoparticles. Chem. Commun. 2019;55(49):6964–6996. doi: 10.1039/C9CC01741K. [DOI] [PubMed] [Google Scholar]
  51. Yanilkin V. V., Nasretdinova G. R., Kokorekin V. A.. Mediated Electrochemical Synthesis of Metal Nanoparticles. Russ. Chem. Rev. 2018;87(11):1080–1110. doi: 10.1070/RCR4827. [DOI] [Google Scholar]
  52. Flores-Rojas G. G., López-Saucedo F., Bucio E.. Gamma-Irradiation Applied in the Synthesis of Metallic and Organic Nanoparticles: A Short Review. Radiat. Phys. Chem. 2020;169:107962. doi: 10.1016/j.radphyschem.2018.08.011. [DOI] [Google Scholar]
  53. Čubová K., Čuba V.. Synthesis of Inorganic Nanoparticles by Ionizing Radiation – a Review. Radiat. Phys. Chem. 2020;169:108774. doi: 10.1016/j.radphyschem.2020.108774. [DOI] [Google Scholar]
  54. Turkevich J., Stevenson P. C., Hillier J.. A Study of the Nucleation and Growth Processes in the Synthesis of Colloidal Gold. Discuss. Faraday Soc. 1951;11(0):55–75. doi: 10.1039/df9511100055. [DOI] [Google Scholar]
  55. Brust M., Walker M., Bethell D., Schiffrin D. J., Whyman R.. Synthesis of Thiol-Derivatised Gold Nanoparticles in a Two-Phase Liquid–Liquid System. J. Chem. Soc. Chem. Commun. 1994;0(No. 7):801–802. doi: 10.1039/C39940000801. [DOI] [Google Scholar]
  56. Uson L., Sebastian V., Arruebo M., Santamaria J.. Continuous Microfluidic Synthesis and Functionalization of Gold Nanorods. Chem. Eng. J. 2016;285:286–292. doi: 10.1016/j.cej.2015.09.103. [DOI] [Google Scholar]
  57. Khan Z., Singh T., Hussain J. I., Hashmi A. A.. Au­(III)–CTAB Reduction by Ascorbic Acid: Preparation and Characterization of Gold Nanoparticles. Colloids Surf., B. 2013;104:11–17. doi: 10.1016/j.colsurfb.2012.11.017. [DOI] [PubMed] [Google Scholar]
  58. Boca S. C., Potara M., Toderas F., Stephan O., Baldeck P. L., Astilean S.. Uptake and Biological Effects of Chitosan-Capped Gold Nanoparticles on Chinese Hamster Ovary Cells. Mater. Sci. Eng.: C. 2011;31(2):184–189. doi: 10.1016/j.msec.2010.08.015. [DOI] [Google Scholar]
  59. Zhao L., Jiang D., Cai Y., Ji X., Xie R., Yang W.. Tuning the Size of Gold Nanoparticles in the Citrate Reduction by Chloride Ions. Nanoscale. 2012;4(16):5071–5076. doi: 10.1039/c2nr30957b. [DOI] [PubMed] [Google Scholar]
  60. Wang M., Sun C., Wang L., Ji X., Bai Y., Li T., Li J.. Electrochemical Detection of DNA Immobilized on Gold Colloid Particles Modified Self-Assembled Monolayer Electrode with Silver Nanoparticle Label. J. Pharm. Biomed. Anal. 2003;33(5):1117–1125. doi: 10.1016/S0731-7085(03)00411-4. [DOI] [PubMed] [Google Scholar]
  61. Costa, D. C. ; Fernandes, M. ; Moura, C. ; Miranda, G. ; Silva, F. ; Carvalho, Ó. ; Madeira, S. . Laser Ablation in Liquid-Assisted Synthesis of Three Types of Nanoparticles for Enhanced Antibacterial Applications Int. J. Precis. Eng. Manuf. - Green Technol. 2025. 10.1007/s40684-025-00711-6. [DOI]
  62. ALmomani A. S., Omar A. F., Oglat A. A., Al-Mafarjy S. S., Dheyab M. A., Khazaalah T. H.. Developed a Unique Technique for Creating Stable Gold Nanoparticles (AuNPs) to Explore Their Potential against Cancer. S. Afr. J. Chem. Eng. 2025;53:142–152. doi: 10.1016/j.sajce.2025.04.012. [DOI] [Google Scholar]
  63. Khlaief, J. H. ; Nayef, U. M. ; AbdulHussien, A. M. . Synthesis of Au@Fe3O4 (Core@Shell) Nanoparticles via Laser Ablation Deposited on Porous Silicon for Photodetector Applications Plasmonics 2025. 10.1007/s11468-025-03036-x. [DOI]
  64. Ali H. A., Abdulhadi O. O., Rzaij J. M.. The Effect of Synthesized Gold Nanoparticles by Laser Ablation Method on Levels of Creatine Kinase Activity. Lasers Manuf. Mater. Process. 2025;12:86–97. doi: 10.1007/s40516-024-00274-5. [DOI] [Google Scholar]
  65. Aziz J., Alfahed R. K. F., Zainulabdeen F. S.. Effect of Gold Doping via Laser Ablation on the Structural, Morphologic, and Optical Properties of CdS Thin Films for Use in Optoelectronic Applications. J. Mater. Sci.: Mater. Electron. 2025;36(12):713. doi: 10.1007/s10854-025-14811-1. [DOI] [Google Scholar]
  66. Rasheed, S. A. ; Nayef, U. M. . Enhancing of Spectral Responsivity of Gold Nanoparticles via Laser Ablation Deposited on Porous Silicon Plasmonics 2025. 10.1007/s11468-025-02821-y. [DOI]
  67. Khlaief, J. H. ; Nayef, U. M. ; Hussien, A. M. A. . Enhancing the Spectral Responsivity of Au@CuO (Core@shell) Nanoparticles via Laser Ablation Deposited on Porous Silicon Plasmonics 2025. 10.1007/s11468-025-02949-x. [DOI]
  68. Rashid, T. M. ; Rahmah, M. I. ; Mahmood, W. K. ; Fahem, M. Q. ; Jabir, M. S. ; Bidan, A. K. ; Adbalrazaq, S. ; Jawad, M. H. ; Awaid, D. M. ; Qamandar, M. A. ; Alsaffar, S. M. . Eco-Friendly Laser Ablation for Synthesis of CNF@Au Nanoparticles: Insights into Enhancing NO2 Gas Detection and Antibacterial Activity Plasmonics 2025. 10.1007/s11468-025-02874-z. [DOI]
  69. Rasheed, S. A. ; Nayef, U. M. . Effect of Laser Energy on Formation of Gold@Aluminum Oxide (Core@Shell) Nanoparticles for Enhancement Spectral Responsivity Plasmonics 2025. 10.1007/s11468-025-02842-7. [DOI]
  70. Al-Kattan A., Tselikov G., Metwally K., Popov A. A., Mensah S., Kabashin A. V., Kulinich S.. Laser Ablation-Assisted Synthesis of Plasmonic Si@Au Core-Satellite Nanocomposites for Biomedical Applications. Nanomaterials. 2021;11:592. doi: 10.3390/nano1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kresnik L., Majerič P., Feizpour D., Črešnar K. P., Rudolf R.. Hybrid Nanostructures of Fe3O4 and Au Prepared via Coprecipitation and Ultrasonic Spray Pyrolysis. Metals. 2024;14(12):1324. doi: 10.3390/met14121324. [DOI] [Google Scholar]
  72. Jelen Ž., Majerič P., Zadravec M., Anzel I., Rakuša M., Rudolf R.. Study of Gold Nanoparticles’ Preparation through Ultrasonic Spray Pyrolysis and Lyophilisation for Possible Use as Markers in LFIA Tests. Nanotechnol. Rev. 2021;10(1):1978–1992. doi: 10.1515/ntrev-2021-0120. [DOI] [Google Scholar]
  73. Soden D., Taqy S., Ghosh K., Haque A.. A Tunable Approach to Fabricate Cost-Effective SERS Substrates Using Au Nanoparticles by Sputtering Deposition. Bull. Mater. Sci. 2025;48(1):30. doi: 10.1007/s12034-024-03395-x. [DOI] [Google Scholar]
  74. Mukherjee D., Kertész K., Zolnai Z., Kovács Z., Deák A., Pálinkás A., Osváth Z., Olasz D., Romanenko A., Fried M., Burger S., Sáfrán G., Petrik P.. Optimized Sensing on Gold Nanoparticles Created by Graded-Layer Magnetron Sputtering and Annealing. Sens Actuators, B. 2025;425:136875. doi: 10.1016/j.snb.2024.136875. [DOI] [Google Scholar]
  75. Lamtha T., Waiwijit U., Duangkanya K., Lertvanithphol T., Amarit R., Tantiwanichapan K., Sathukarn A., Chananonnawathorn C., Hincheeranan W., Dhanasiwawong K., Choowongkomon K., Sritrakul T., Boodde O., Sukmak M., Horprathum M.. Solid-State Dewetting Sputtered Ultra-Thin Au Films for LSPR Sensing Chip toward African Swine Fever Virus Detection. Sens Actuators, A. 2025;382:116165. doi: 10.1016/j.sna.2024.116165. [DOI] [Google Scholar]
  76. Besner S., Kabashin A. V., Winnik F. M., Meunier M.. Synthesis of Size-Tunable Polymer-Protected Gold Nanoparticles by Femtosecond Laser-Based Ablation and Seed Growth. J. Phys. Chem. C. 2009;113(22):9526–9531. doi: 10.1021/jp809275v. [DOI] [Google Scholar]
  77. Pokhrel Y., Tack M., Levantino M., Reichenberger S., Plech A.. Caging Effect of Unbound Graphene Oxide Nanosheets during Gold Colloid Fragmentation under Electrostatic Repulsion. J. Phys. Chem. C. 2025;129:8252–8261. doi: 10.1021/acs.jpcc.5c00439. [DOI] [Google Scholar]
  78. Hettiarachchi B. S., Yakiyama Y., Sakurai H.. Tailoring Carbon-Encapsulated Gold Nanoclusters via Microchip Laser Ablation in Polystyrene Solution: Controlling Size, Structure, and Photoluminescent Properties. RSC Appl. Interfaces. 2025;2:772–779. doi: 10.1039/D4LF00349G. [DOI] [Google Scholar]
  79. Shbeeb, R. T. ; Mutlak, F. A. H. . Effective Performance of Gold–Zirconia Photodetector Produced via Laser Energy Control Deposited on Porous Silicon Plasmonics 2025. 10.1007/s11468-025-03039-8. [DOI]
  80. Salim, E. T. ; Mahdi, R. O. ; Mahmoud, D. ; Gopinath, S. C. B. ; Qaeed, M. A. . Effect of Laser Wavelength on the Structural, Morphological, and Optical Plasmonic Properties of Au@WO3 Core–Shell NPs Plasmonics 2025. 10.1007/s11468-025-02929-1. [DOI]
  81. Figat A. M., Bartosewicz B., Liszewska M., Budner B., Norek M., Jankiewicz B. J.. α-Amino Acids as Reducing and Capping Agents in Gold Nanoparticles Synthesis Using the Turkevich Method. Langmuir. 2023;39(25):8646–8657. doi: 10.1021/acs.langmuir.3c00507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Pothukuchi R. P., Kumar S., Punnathanam S. N.. Mechanistic Details of the Organizer-Assisted Nucleation of Gold Nanoparticles by the Turkevich Method. J. Phys. Chem. C. 2024;128:17642–17650. doi: 10.1021/acs.jpcc.4c03537. [DOI] [Google Scholar]
  83. Bilal M., Bandyopadhyay S.. Controlled Growth of Citrate-Stabilized Gold Nanoparticles Using a Semi-Continuous Seed-Mediated Route. Discover Nano. 2025;20(1):39. doi: 10.1186/s11671-025-04189-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Idar I., Yusuf M., Al Anshori J., Subroto T.. IgY Anti-N SARS CoV2-Conjugated Gold Nanoparticles Production for Lateral Flow Biosensor-Based Diagnostics. Trends Sci. 2025;22(4):9466. doi: 10.48048/tis.2025.9466. [DOI] [Google Scholar]
  85. Liu H., Li Y., Li T., Mu Y., Fang X., Zhang X.. Mono-, Di- and Trimetallic Coinage Nanoparticles Prepared via the Brust-Schiffrin Method. Phys. Chem. Chem. Phys. 2024;26(25):17760–17768. doi: 10.1039/D4CP01530D. [DOI] [PubMed] [Google Scholar]
  86. Kang D., Son C., Lee H., Park H., Kim T., Park J.. Aqueous Synthesis of the Tiopronin-Capped Gold Nanoclusters/Nanoparticles with Precise Size Control via Deprotonation of the Ligand. Appl. Sci. 2022;12(16):8263. doi: 10.3390/app12168263. [DOI] [Google Scholar]
  87. Wagner M., Herrero-Ruiz A., Verde-Sesto E., Asenjo-Sanz I., Liz-Marzán L. M.. Influence of Poly­(Vinylpyrrolidone) Synthesis Conditions on the Formation of Gold Nanostars. Chem. Mater. 2025;37:644–654. doi: 10.1021/acs.chemmater.4c02513. [DOI] [Google Scholar]
  88. Khan N. A., Kuo Y. H., Yu S. Y., Ku Y. C., Hung K. Y., Liaw J. W.. Laser-Induced Photochemical Reduction for the Production of Gold and Silver Nanoparticles on Gold Nanostructures. Results Phys. 2025;70:108152. doi: 10.1016/j.rinp.2025.108152. [DOI] [Google Scholar]
  89. Choleva T. G., Karagianni V. I., Giokas D. L.. Hydrochromic Paper-Based Dosimeter for Monitoring UV Light Exposure Based on the Photochemical Formation of Gold Nanoparticles. Microchim. Acta. 2025;192(3):169. doi: 10.1007/s00604-025-07020-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Im S. W., Kim R. M., Han J. H., Ha I. H., Lee H. E., Ahn H. Y., Jo E., Nam K. T.. Synthesis of Chiral Gold Helicoid Nanoparticles Using Glutathione. Nat. Protoc. 2025;20:1082–1096. doi: 10.1038/s41596-024-01083-y. [DOI] [PubMed] [Google Scholar]
  91. D P. N., Sinha R. K.. Shape-Controlled Synthesis and Bulk Refractive Index Sensitivity Studies of Gold Nanoparticles for LSPR-Based Sensing. Plasmonics. 2024;20:1351–1364. doi: 10.1007/s11468-024-02387-1. [DOI] [Google Scholar]
  92. Panagopoulos D., Alamdari A. A., Quinson J.. Surfactant-Free Colloidal Gold Nanoparticles: Room Temperature Synthesis, Size Control and Opportunities for Catalysis. Mater. Today Nano. 2025;29:100600. doi: 10.1016/j.mtnano.2025.100600. [DOI] [Google Scholar]
  93. Oliveira A. E. F., Pereira A. C., Resende M. A. C., Ferreira L. F.. Gold Nanoparticles: A Didactic Step-by-Step of the Synthesis Using the Turkevich Method, Mechanisms, and Characterizations. Analytica. 2023;4(2):250–263. doi: 10.3390/analytica4020020. [DOI] [Google Scholar]
  94. Bilal M., Bandyopadhyay S.. Controlled Growth of Citrate-Stabilized Gold Nanoparticles Using a Semi-Continuous Seed-Mediated Route. Discover Nano. 2025;20(1):39. doi: 10.1186/s11671-025-04189-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Mattioni, J. V. ; da Silva, L. K. A. ; Silva, H. N. ; Franciscato, D. S. ; Toma, H. E. . PlasmonicsShort Communication Do Nanoparticles’Lability Influence Plasmonics and SERS? Plasmonics 2025. 10.1007/s11468-025-03021-4. [DOI]
  96. Souza L. H. M., Dias I. R., von Zuben de Valega Negrão C., da Costa Oliveira H., Turrini P. C. G., da Costa Martins A. G., dos Reis J. L. M. S., Yepez E. A. V., Gobara B. N. K., Perecin C. J., Pesquero J. B., Verona B. M., Cerize N. N. P.. Gold Nanoparticle-DNA Conjugates: An Enzymatic DNA Synthesis Platform. ACS Omega. 2025;10:20452–50464. doi: 10.1021/acsomega.5c00645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Paul D., Ganesan R., Ray Dutta J.. Effect of Polymeric Additives on Gold Nanoparticles for Colorimetric Detection of Hepatitis C Virus. Gold Bull. 2025;58(1):8. doi: 10.1007/s13404-025-00362-0. [DOI] [Google Scholar]
  98. Duggal M., Khatavi S., Yelamaggad C. V., Rao D. S. S., Prasad S. K., Prasad B. L. V.. Vastly Increased Mesophase Range Due to Superstructure Formation from In Situ Prepared Gold Nanoparticles with Liquid Crystalline Ligands: Toward Enhanced Optoelectronic Applications. ACS Appl. Nano Mater. 2025;8:10340–10350. doi: 10.1021/acsanm.5c00923. [DOI] [Google Scholar]
  99. Wagner L. S., Thiele T., Loza K., Beuck C., Bayer P., Heggen M., Giese M., Epple M.. Ultrasmall Gold Nanoparticles (2 Nm) Decorated with a High Density of Photochemically Switchable Ligands. Chem. - Eur. J. 2025;31:e202501204. doi: 10.1002/chem.202501204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Stolarczyk E. U., Strzempek W., Muszyńska M., Kubiszewski M., Witkowska A. B., Trzcińska K., Wojdasiewicz P., Stolarczyk K.. Preparation of Diosgenin-Functionalized Gold Nanoparticles: From Synthesis to Antitumor Activities. Int. J. Mol. Sci. 2025;26(3):1088. doi: 10.3390/ijms26031088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Verma J., Kumar C., Sharma M., Saxena S.. Biotechnological Advances in Microbial Synthesis of Gold Nanoparticles: Optimizations and Applications. 3 Biotech. 2024;14:263. doi: 10.1007/s13205-024-04110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Mehra V., Kumar S., Tamang A. M., Chandraker S. K.. Green Synthesis of Gold Nanoparticles (AuNPs) by Using Plant Extract and Their Biological Application: A Review. BioNanoScience. 2025;15:18. doi: 10.1007/s12668-024-01703-7. [DOI] [Google Scholar]
  103. Pathan A., Nayak T., Alshahrani S., Tripathi R., Tripathi P.. Current and Emerging Frontiers in Biologically Synthesized Gold Nanoparticles: An in-Depth Review. Chem. Pap. 2025;79:3421–3442. doi: 10.1007/s11696-025-03952-6. [DOI] [Google Scholar]
  104. Sheershwal A., Singh A., Sharma V., Trivedi B.. Microbial Synthesis of Nanoparticles for Sustainable Agricultural Advancements: A Comprehensive Review. Nanotechno. Environ. Eng. 2025;10(1):16. doi: 10.1007/s41204-025-00405-z. [DOI] [Google Scholar]
  105. Nithin B. R., Bhuyar P., Maniam G. P., Rahim M. H. A., Govindan N.. Environment Friendly Approach for Plant Mediated Green Biosynthesis of Gold Nanoparticles and Their Modern Applications in Biomedical Aspectsan Updated Report. BioNanoScience. 2023;13:1517–1540. doi: 10.1007/s12668-023-01134-w. [DOI] [Google Scholar]
  106. Siddiqi K. S., Husen A.. Recent Advances in Plant-Mediated Engineered Gold Nanoparticles and Their Application in Biological System. J. Trace Elem. Med. Biol. 2017;40:10–23. doi: 10.1016/j.jtemb.2016.11.012. [DOI] [PubMed] [Google Scholar]
  107. Srinath B. S., Namratha K., Byrappa K.. Eco-Friendly Synthesis of Gold Nanoparticles by Gold Mine Bacteria Brevibacillus Formosus and Their Antibacterial and Biocompatible Studies. IOSR J. Pharm. 2017;7:53–60. [Google Scholar]
  108. Singh P. K., Kundu S.. Biosynthesis of Gold Nanoparticles Using Bacteria. Proc. Natl. Acad. Sci., India, Sect. B. 2014;84(2):331–336. doi: 10.1007/s40011-013-0230-6. [DOI] [Google Scholar]
  109. Altaf I., Firdaus N., Wani I. A., Khan S., Owais M.. Iris Kashmiriana Mediated Synthesis of Gold Nanoparticles: Characterization and Its Antibacterial Implications on Dental Appliances. Bionanoscience. 2025;15(1):133. doi: 10.1007/s12668-024-01693-6. [DOI] [Google Scholar]
  110. Daramola O. B., Torimiro N., George R. C.. Colorimetric-Based Detection of Enteric Bacterial Pathogens Using Chromogens-Functionalized Iron Oxide-Gold Nanocomposites Biosynthesized by Bacillus Subtilis. Discover Biotechnol. 2025;2(1):1. doi: 10.1007/s44340-025-00008-z. [DOI] [Google Scholar]
  111. Abu-Elghait M., Soliman M. K. Y., Azab M. S., Salem S. S.. Response Surface Methodology: Optimization of Myco-Synthesized Gold and Silver Nanoparticles by Trichoderma Saturnisporum. Biomass Convers. Biorefin. 2025;15:4211–4224. doi: 10.1007/s13399-023-05188-4. [DOI] [Google Scholar]
  112. Abed A. S., Khalaf Y. H., Mohammed A. M.. Green Synthesis of Gold Nanoparticles as an Effective Opportunity for Cancer Treatment. Results Chem. 2023;5:100848. doi: 10.1016/j.rechem.2023.100848. [DOI] [Google Scholar]
  113. Patil T. P., Vibhute A. A., Patil S. L., Dongale T. D., Tiwari A. P.. Green Synthesis of Gold Nanoparticles via Capsicum Annum Fruit Extract: Characterization, Antiangiogenic, Antioxidant and Anti-Inflammatory Activities. Appl. Surf. Sci. Adv. 2023;13:100372. doi: 10.1016/j.apsadv.2023.100372. [DOI] [Google Scholar]
  114. Shabani L., Kasaee S. R., Chelliapan S., Abbasi M., Khajehzadeh H., Dehghani F. S., Firuzyar T., Shafiee M., Amani A. M., Mosleh-Shirazi S., Vaez A., Kamyab H.. An Investigation into Green Synthesis of Ru Template Gold Nanoparticles and the in Vitro Photothermal Effect on the MCF-7 Human Breast Cancer Cell Line. Appl. Phys. A. 2023;129(8):564. doi: 10.1007/s00339-023-06832-6. [DOI] [Google Scholar]
  115. Moosavy M. H., de la Guardia M., Mokhtarzadeh A., Khatibi S. A., Hosseinzadeh N., Hajipour N.. Green Synthesis, Characterization, and Biological Evaluation of Gold and Silver Nanoparticles Using Mentha Spicata Essential Oil. Sci. Rep. 2023;13(1):7320. doi: 10.1038/s41598-023-33632-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Ren Z., Li H., Li J., Cai J., Zhong L., Ma Y., Pang Y.. Green Synthesis of Reduced Graphene Oxide/Chitosan/Gold Nanoparticles Composites and Their Catalytic Activity for Reduction of 4-Nitrophenol. Int. J. Biol. Macromol. 2023;229:732–745. doi: 10.1016/j.ijbiomac.2022.12.282. [DOI] [PubMed] [Google Scholar]
  117. Palaniyandi T., Viswanathan S., Prabhakaran P., Baskar G., Wahab M. R. A., Sivaji A., Ravi M., Rajendran B. K., Moovendhan M., Surendran H., Kumarasamy S.. Green Synthesis of Gold Nanoparticles Using Halymenia Pseudofloresii Extracts and Their Antioxidant, Antimicrobial, and Anti-Cancer Activities. Biomass Convers. Biorefin. 2023 doi: 10.1007/s13399-023-03873-y. [DOI] [Google Scholar]
  118. Akkam N., Aljabali A. A. A., Akkam Y., Abo Alrob O., Al-Trad B., Alzoubi H., Tambuwala M. M., Al-Batayneh K. M.. Investigating the Fate and Toxicity of Green Synthesized Gold Nanoparticles in Albino Mice. Drug Dev. Ind. Pharm. 2023;49(8):508–520. doi: 10.1080/03639045.2023.2243334. [DOI] [PubMed] [Google Scholar]
  119. Nardi N., Baumgarten L. G., Dreyer J. P., Santana E. R., Winiarski J. P., Vieira I. C.. Nanocomposite Based on Green Synthesis of Gold Nanoparticles Decorated with Functionalized Multi-Walled Carbon Nanotubes for the Electrochemical Determination of Hydroxychloroquine. J. Pharm. Biomed. Anal. 2023;236:115681. doi: 10.1016/j.jpba.2023.115681. [DOI] [PubMed] [Google Scholar]
  120. Do Dat T., Cong C. Q., Le Hoai Nhi T., Khang P. T., Nam N. T. H., Thi Tinh N., Hue D. T., Hieu N. H.. Green Synthesis of Gold Nanoparticles Using Andrographis Paniculata Leave Extract for Lead Ion Detection, Degradation of Dyes, and Bioactivities. Biochem. Eng. J. 2023;200:109103. doi: 10.1016/j.bej.2023.109103. [DOI] [Google Scholar]
  121. Dehghani F., Mosleh-Shirazi S., Shafiee M., Kasaee S. R., Amani A. M.. Antiviral and Antioxidant Properties of Green Synthesized Gold Nanoparticles Using Glaucium Flavum Leaf Extract. Appl. Nanosci. 2023;13(6):4395–4405. doi: 10.1007/s13204-022-02705-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Al-Radadi N. S.. Ephedra Mediated Green Synthesis of Gold Nanoparticles (AuNPs) and Evaluation of Its Antioxidant, Antipyretic, Anti-Asthmatic, and Antimicrobial Properties. Arabian J. Chem. 2023;16(1):104353. doi: 10.1016/j.arabjc.2022.104353. [DOI] [Google Scholar]
  123. Worakitjaroenphon, S. ; Shanmugam, P. ; Boonyuen, S. ; Smith, S. M. ; Chookamnerd, K. . Green Synthesis of Silver and Gold Nanoparticles Using Oroxylum Indicum Plant Extract for Catalytic and Antimicrobial Activity Biomass Convers. Biorefin. 2023. 10.1007/s13399-023-04734-4. [DOI]
  124. Raziani Y., Shakib P., Rashidipour M., Cheraghipour K., Yadegari J. G., Mahmoudvand H.. Green Synthesis, Characterization, and Antiparasitic Effects of Gold Nanoparticles against Echinococcus Granulosus Protoscoleces. Trop. Med. Infect. Dis. 2023;8(6):313. doi: 10.3390/tropicalmed8060313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Döll E. G., Santana E. R., Winiarski J. P., Baumgarten L. G., Vieira I. C.. Green Synthesis of Gold Nanoparticles Using Peach Extract Incorporated in Graphene for the Electrochemical Determination of Antioxidant Butylated Hydroxyanisole in Food Matrices. Biosensors. 2023;13(12):1037. doi: 10.3390/bios13121037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Yaou Balarabe B.. Green Synthesis of Gold-Titania Nanoparticles for Sustainable Ciprofloxacin Removal and Phytotoxicity Evaluation on Aquatic Plant Growth. Hybrid Adv. 2023;4:100107. doi: 10.1016/j.hybadv.2023.100107. [DOI] [Google Scholar]
  127. Hamelian M., Varmira K., Karmakar B., Veisi H.. Catalytic Reduction of 4-Nitrophenol Using Green Synthesized Silver and Gold Nanoparticles over Thyme Plant Extract. Catal. Lett. 2023;153(8):2341–2351. doi: 10.1007/s10562-022-04164-3. [DOI] [Google Scholar]
  128. Ashikbayeva Z., Bekmurzayeva A., Myrkhiyeva Z., Assylbekova N., Atabaev T. S., Tosi D.. Green-Synthesized Gold Nanoparticle-Based Optical Fiber Ball Resonator Biosensor for Cancer Biomarker Detection. Opt. Laser Technol. 2023;161:109136. doi: 10.1016/j.optlastec.2023.109136. [DOI] [Google Scholar]
  129. Faid A. H., Shouman S. A., Thabet N. A., Badr Y. A., Sliem M. A.. Laser Enhanced Combinatorial Chemo-Photothermal Therapy of Green Synthesis Gold Nanoparticles Loaded with 6Mercaptopurine on Breast Cancer Model. J. Pharm. Innovation. 2023;18(1):144–148. doi: 10.1007/s12247-022-09626-0. [DOI] [Google Scholar]
  130. Umamaheswari K., Abirami M.. Assessment of Antifungal Action Mechanism of Green Synthesized Gold Nanoparticles (AuNPs) Using Allium Sativum on Candida Species. Mater. Lett. 2023;333:133616. doi: 10.1016/j.matlet.2022.133616. [DOI] [Google Scholar]
  131. Rokkarukala S., Cherian T., Ragavendran C., Mohanraju R., Kamaraj C., Almoshari Y., Albariqi A., Sultan M. H., Alsalhi A., Mohan S.. One-Pot Green Synthesis of Gold Nanoparticles Using Sarcophyton Crassocaule, a Marine Soft Coral: Assessing Biological Potentialities of Antibacterial, Antioxidant, Anti-Diabetic and Catalytic Degradation of Toxic Organic Pollutants. Heliyon. 2023;9(3):e14668. doi: 10.1016/j.heliyon.2023.e14668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Wang L., Qiang X., Song Y., Wang X., Gu W., Niu J., Sun Y., Srinuanpan S., Wang G.. Green Synthesis of Gold Nanoparticles by Phycoerythrin Extracted from Solieria Tenuis as an Efficient Catalyst for 4-Nitrophenol Reduction and Degradation of Dyes in Wastewater. Mater. Today Sustainability. 2023;23:100435. doi: 10.1016/j.mtsust.2023.100435. [DOI] [Google Scholar]
  133. Torres-Ortiz D., García-Alcocer G., Loske A. M., Fernández F., Becerra-Becerra E., Esparza R., Gonzalez-Reyna M. A., Estevez M.. Green Synthesis and Antiproliferative Activity of Gold Nanoparticles of a Controlled Size and Shape Obtained Using Shock Wave Extracts from Amphipterygium Adstringens. Bioengineering. 2023;10(4):437. doi: 10.3390/bioengineering10040437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Velidandi A., Sarvepalli M., Gandam P. K., Baadhe R. R.. Silver/Silver Chloride and Gold Bimetallic Nanoparticles: Green Synthesis Using Azadirachta Indica Aqueous Leaf Extract, Characterization, Antibacterial, Catalytic, and Recyclability Studies. Inorg. Chem. Commun. 2023;155:111107. doi: 10.1016/j.inoche.2023.111107. [DOI] [Google Scholar]
  135. Ramli M., Sujoko I., Adhha N., Annas D., Nikmatullah M., Ariyanta H. A., Apriandanu D. O. B., Saputra I. S., Khairurrijal K.. Influence of Various Concentrations of Chloroauric Acid on the Fabrication of Gold Nanoparticles: Green Synthesis Using Elaeis Guineensis Jacq Leaf Extract and Characterizations. Results Surf. Interfaces. 2024;17:100317. doi: 10.1016/j.rsurfi.2024.100317. [DOI] [Google Scholar]
  136. Al-Mafarjy S. S., Suardi N., Ahmed N. M., Kernain D., Alkatib H. H., Dheyab M. A.. Green Synthesis of Gold Nanoparticles from Coleus Scutellarioides (L.) Benth Leaves and Assessment of Anticancer and Antioxidant Properties. Inorg. Chem. Commun. 2024;161:112052. doi: 10.1016/j.inoche.2024.112052. [DOI] [Google Scholar]
  137. İpek P., Baran M. F., Baran A., Hatipoğlu A., Keskin C., Yildiztekin M., Küçükaydin S., Becerekli H., Kurt K., Eftekhari A., Huseynova I., Khalilov R.. Cho, W. C. Green Synthesis and Evaluation of Antipathogenic, Antioxidant, and Anticholinesterase Activities of Gold Nanoparticles (Au NPs) from Allium Cepa L. Peel Aqueous Extract. Biomass Convers. Biorefin. 2024;14(9):10661–10670. doi: 10.1007/s13399-023-04362-y. [DOI] [Google Scholar]
  138. Oliveira M., Sousa A., Sá S., Soares S., Pereira A. C., Rocha A. C., Pais P., Ferreira D., Almeida C., Luís C., Lima C., Almeida F., Gestoso Á., Duarte M. C., Barata P., Martins-Mendes D., Baylina P., Pereira C. F., Fernandes R.. Harvesting the Power of Green Synthesis: Gold Nanoparticles Tailored for Prostate Cancer Therapy. Int. J. Mol. Sci. 2024;25(4):2277. doi: 10.3390/ijms25042277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Bohdanovych T., Kuzema P., Anishchenko V., Duplij V., Kharchuk M., Lyzhniuk V., Shakhovsky A., Matvieieva N.. Comparison of Silver and Gold Nanoparticles Green Synthesis by Artemisia Annua Hairy Root Extracts. Biol. Open. 2025;14(3):bio061739. doi: 10.1242/bio.061739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Lavanya G., Anandaraj K., Selvam K., Gopu M., Selvankumar T., Govarthanan M., Kumar P.. Green Synthesis of Gold Nanoparticles Using Macroalgae Halimeda Macroloba Extract and Their Photocatalytic Degradation of Methylene Blue and Methyl Orange. Polym. Adv. Technol. 2024;35(4):e6383. doi: 10.1002/pat.6383. [DOI] [Google Scholar]
  141. Ali M. Y., Abdulrahman H. B., Ting W.-T., Howlader M. M. R.. Green Synthesized Gold Nanoparticles and CuO-Based Nonenzymatic Sensor for Saliva Glucose Monitoring. RSC Adv. 2024;14(1):577–588. doi: 10.1039/D3RA05644A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Ullah I., Rauf A., Khalil A. A., Luqman M., Islam M. R., Hemeg H. A., Ahmad Z., Al-Awthan Y. S., Bahattab O., Quradha M. M., Peganum Harmala L.. Extract-Based Gold (Au) and Silver (Ag) Nanoparticles (NPs): Green Synthesis, Characterization, and Assessment of Antibacterial and Antifungal Properties. Food Sci. Nutr. 2024;12(6):4459–4472. doi: 10.1002/fsn3.4112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Al-Radadi N. S., Al-Bishri W. M., Salem N. A., ElShebiney S. A.. Plant-Mediated Green Synthesis of Gold Nanoparticles Using an Aqueous Extract of Passiflora Ligularis, Optimization, Characterizations, and Their Neuroprotective Effect on Propionic Acid-Induced Autism in Wistar Rats. Saudi Pharm. J. 2024;32(2):101921. doi: 10.1016/j.jsps.2023.101921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Zannotti M., Piras S., Remia L., Appignanesi D., Giovannetti R.. Selective Colorimetric Detection of Pb­(II) Ions by Using Green Synthesized Gold Nanoparticles with Orange Peel Extract. Chemosensors. 2024;12(3):33. doi: 10.3390/chemosensors12030033. [DOI] [Google Scholar]
  145. Khan A. W., Lali N. S., Sabei F. Y., Irfan M. I., Naeem-ul-Hassan M., Sher M., Safhi A. Y., Alsalhi A., Albariqi A. H., Kamli F., Amin H. M. A., Abbas A.. Sunlight-Assisted Green Synthesis of Gold Nanocubes Using Horsetail Leaf Extract: A Highly Selective Colorimetric Sensor for Pb2+, Photocatalytic and Antimicrobial Agent. J. Environ. Chem. Eng. 2024;12(3):112576. doi: 10.1016/j.jece.2024.112576. [DOI] [Google Scholar]
  146. Jakhmola A., Hornsby T. K., Kashkooli F. M., Kolios M. C., Rod K., Tavakkoli J.. Green Synthesis of Anti-Cancer Drug-Loaded Gold Nanoparticles for Low-Intensity Pulsed Ultrasound Targeted Drug Release. Drug Delivery Transl. Res. 2024;14(9):2417–2432. doi: 10.1007/s13346-024-01516-x. [DOI] [PubMed] [Google Scholar]
  147. Kabak B., Trak D., Kendüzler E., Arslan Y.. Novel Approach for Green Synthesis of Stable Gold Nanoparticles with Dried Turmeric Root Extract: Investigations on Sensor and Catalytic Applications. Environ. Sci. Pollut. Res. 2024;31(38):50614–50629. doi: 10.1007/s11356-024-34588-w. [DOI] [PubMed] [Google Scholar]
  148. Khalil M. T., Zhang P., Han G., Wu X., Li B., Xiao M.. Green Synthesis of Gold Nanoparticles for Catalytic Reduction of 4-Nitrophenol and Methylene Blue for Sustainable Development. Sustainable Polym. Energy. 2023;1(1):10002. doi: 10.35534/spe.2024.10002. [DOI] [Google Scholar]
  149. Ahmad S., Ahmad S., Xu Q., Khan I., Cao X., Yang R., Yan H.. Green Synthesis of Gold and Silver Nanoparticles Using Crude Extract of Aconitum Violaceum and Evaluation of Their Antibacterial, Antioxidant and Photocatalytic Activities. Front Bioeng. Biotechnol. 2024;11:1320739. doi: 10.3389/fbioe.2023.1320739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Mary S. J., Veeravarmal V., Thankappan P., Arumugam P., Augustine P. I., Franklin R.. Anti-Cancer Effects of Green Synthesized Gold Nanoparticles Using Leaf Extract of Annona Muricata. L against Squamous Cell Carcinoma Cell Line 15 through Apoptotic Pathway. Dent. Res. J. 2024;21:14. doi: 10.4103/drj.drj_521_23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Peng H., Zhang S., Chai Q., Hua Z.. Green Synthesis of Gold Nanoparticles Using Acorus Calamus Leaf Extract and Study on Their Anti-Alzheimer Potential. Biotechnol. Bioprocess Eng. 2024;29(1):157–163. doi: 10.1007/s12257-024-00010-y. [DOI] [Google Scholar]
  152. Adem Z., Bekana D., Temesgen A., Teju E., Amde M., Jabesa A.. Plasmon-Based Colorimetric Assay Using Green Synthesized Gold Nanoparticles for the Detection of Bisphenol A. Anal. Sci. 2024;40(4):671–679. doi: 10.1007/s44211-023-00500-8. [DOI] [PubMed] [Google Scholar]
  153. González-Ballesteros N., Martins P. M., Tavares C. J., Lanceros-Méndez S.. Quercetin-Mediated Green Synthesis of Au/TiO2 Nanocomposites for the Photocatalytic Degradation of Antibiotic Ciprofloxacin. J. Ind. Eng. Chem. 2025;143:526–537. doi: 10.1016/j.jiec.2024.09.003. [DOI] [Google Scholar]
  154. Serdar G.. Gold Nanoparticles Biosynthesis Through Green Synthesis Mediated by Leaf Extract from Diospyros Kaki L. (Persimmon) Using the Microwave Extraction Method. Plasmonics. 2024;19:2813–2823. doi: 10.1007/s11468-024-02294-5. [DOI] [Google Scholar]
  155. Yu T., Liu L., Liu Y., Yin W.. Synthesis of Gold Nanoparticles Using Ginkgo Biloba Leaf Extract and Its in Vitro Anticancer Effects on Nasopharyngeal Carcinoma Cells. Alexandria Eng. J. 2024;104:614–620. doi: 10.1016/j.aej.2024.07.130. [DOI] [Google Scholar]
  156. Rosyidah A., Purbani D. C., Pratiwi R. D., Muttaqien S. E., Nantapong N., Warsito M. F., Fikri M. N., Ruth F., Gustini N., Syahputra G., Padri M., Noerdjito D. R., Nurkanto A., Afani H.. Eco-Friendly Synthesis of Gold Nanoparticles by Marine Microalgae Synechococcus Moorigangae: Characterization, Antimicrobial, and Antioxidant Properties. Kuwait J. Sci. 2024;51(2):100194. doi: 10.1016/j.kjs.2024.100194. [DOI] [Google Scholar]
  157. Gul A., Baig M. N., Ahmed D., Najam Z., Aslam T., Ali S.. Green Synthesis of Silver Nanoparticles from Spirulina Platensis Extract: Antibacterial and Antioxidant Potential. Bionanoscience. 2024;14:2327–2336. doi: 10.1007/s12668-024-01473-2. [DOI] [Google Scholar]
  158. Wang Y., Chen J., Bo Y., Chen J., Liu Y., Li Z., Yu C., Liu J., Wang S.. Green Synthesis of Au Nanoparticles by Scutellaria Barbata Extract for Chemo-Photothermal Anticancer Therapy. Pharmacol. Res. - Mod. Chin. Med. 2024;13:100536. doi: 10.1016/j.prmcm.2024.100536. [DOI] [Google Scholar]
  159. Ghoreishi S. M., Mortazavi-Derazkola S.. Eco-Friendly Synthesis of Gold Nanoparticles via Tangerine Peel Extract: Unveiling Their Multifaceted Biological and Catalytic Potentials. Heliyon. 2025;11(1):e40104. doi: 10.1016/j.heliyon.2024.e40104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Vorobyova V., Skiba M., Vinnichuk K., Vasyliev G.. Synthesis of Gold Nanoparticles Using Plum Waste Extract with Green Solvents. Sustainable Chem. Environ. 2024;6:100086. doi: 10.1016/j.scenv.2024.100086. [DOI] [Google Scholar]
  161. Kaval U., Hoşgören H.. Biosynthesis, Characterization, and Biomedical Applications of Gold Nanoparticles with Cucurbita Moschata Duchesne Ex Poiret Peel Aqueous Extracts. Molecules. 2024;29(5):923. doi: 10.3390/molecules29050923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Sridharan G., Murugan R. V., Atchudan R., Arya S., Sundramoorthy A. K.. Electrochemical Detection of Dopamine Using Green Synthesized Gold Nanoparticles from Strobilanthes Kunthiana’s Leaf Extract. Nano Life. 2025;15:2450015. doi: 10.1142/S1793984424500156. [DOI] [Google Scholar]
  163. Wehbe N., Mesmar J. E., El Kurdi R., Al-Sawalmih A., Badran A., Patra D., Baydoun E.. Halodule Uninervis Extract Facilitates the Green Synthesis of Gold Nanoparticles with Anticancer Activity. Sci. Rep/ 2025;15(1):4286. doi: 10.1038/s41598-024-81875-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Acquavia M. A., Gubitosa J., Rizzi V., Cosma P., Gaeta C., Iannece P., Della Sala P., Guerrieri A., Tesoro C., Ciriello R., Lelario F., Gregori E., Montoro P., Di Capua A., Bianco G.. Green Synthesis of Gold Nanoparticles from Pomegranate Juice Ascertained by a Combined Approach Based on MALDI FT-ICR MS and LC-ESI-MS/MS. Food Chem. 2025;476:143427. doi: 10.1016/j.foodchem.2025.143427. [DOI] [PubMed] [Google Scholar]
  165. Ávila-Avilés R. D., Becerril-Juárez I. G., Vilchis-Nestor A. R.. In Situ Green Synthesis of Graphene-Based Material Nanocomposites Decorated with Silver and Gold Nanoparticles for Improved SERS Sensing. Opt Mater. 2025;160:116730. doi: 10.1016/j.optmat.2025.116730. [DOI] [Google Scholar]
  166. El-Said W. A., Akhdhar A., Al-Bogami A. S., Saleh T. S.. Design and Green Synthesis of Carbon Dots/Gold Nanoparticles Composites and Their Applications for Neurotransmitters Sensing Based on Emission Spectroscopy. Spectrochim. Acta, Part A. 2025;327:125402. doi: 10.1016/j.saa.2024.125402. [DOI] [PubMed] [Google Scholar]
  167. Saha D., Talukdar D., Mukherjee P., Mitra D., Mukherjee R., Guha S., Bhattacharjee A., Naskar R., Kumar Sahu S., Alam N., Das G., Murmu N.. Green Synthesis of Gold Nano-Particles Using Madhuca Indica Flower Extract and Their Anticancer Activity on Head and Neck Cancer: Characterization and Mechanistic Study. Eur. J. Pharm. Biopharm. 2025;207:114625. doi: 10.1016/j.ejpb.2025.114625. [DOI] [PubMed] [Google Scholar]
  168. Lafta M. Z., Al-Samarrai R. R. H., Bouaziz M.. Green Synthesis of Silver and Gold Nanoparticles Using Quercetin Extracted from Arctium Lappa by HPLC, Characterization and Estimation of Antioxidant Activity. Results Chem. 2025;13:102028. doi: 10.1016/j.rechem.2025.102028. [DOI] [Google Scholar]
  169. Patiño-González M. C., Echeverri-Cuartas C. E., Torijano-Gutiérrez S., Naranjo-Rios S. M., Agudelo N. A.. Optimization of the Green Synthesis of Gold Nanorods Using Aqueous Extract of Peeled Sour Guava as a Source of Antioxidants. PLoS One. 2025;20:e0313485. doi: 10.1371/journal.pone.0313485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Khan I. S., Yanamadala S., Chinnaiyan S., Chiterasu N., Kannan S.. Green Synthesis of Gold Nanoparticles Using Sphaeranthus Amaranthoides: Drug Loading and Anticancer Properties in Nanomedicine. Pharmacognosy Res. 2025;17(1):108–114. doi: 10.5530/pres.20251946. [DOI] [Google Scholar]
  171. Wang, R. ; Ni, W. ; Fang, S. ; Chen, J. ; Fan, X. . Ultrasound Assisted Green Synthesis of Lignosulfonate-Gold Nanocomposite (LS-Au NPs): The Study of Its Application in the Suzuki–Miyaura Coupling Reactions and Its Application for the Treatment of Gastric Cancer J. Inorg. Organomet. Polym. Mater. 2025. 10.1007/s10904-024-03578-3. [DOI]
  172. Elizalde-Mata A., Trejo-Caballero M. E., Molina G. A., España-Sánchez B. L., Estevez M.. Green Synthesis of Gold Nanoparticles from Dragon’s Blood (Croton Draco) Aqueous Extracts for Biomedical Applications. MRS Adv. 2025;10:65–71. doi: 10.1557/s43580-024-01038-1. [DOI] [Google Scholar]
  173. Dewi F. R. P., Wahyuningsih S. P. A., Tan S. C., Lim V., Jamil A. K. M., Yusof N. S. M., Muthu P., Islamatasya U.. Green Synthesis of Gold Nanoparticles from Stachytarpheta Jamaicensis: An Eco-Friendly Approach with Promising Anticancer Potency. Nanomed. J. 2025;12(1):51–58. doi: 10.22038/NMJ.2024.77277.1883. [DOI] [Google Scholar]
  174. Sadeghi-Kiakhani M., Hashemi E., Norouzi M. M., Soleimani P., Babaahmadi V.. Green Synthesis of Silver Nanoparticles on Polyamide Fabrics Using Scrophularia Striata Boiss Extract: Characterization, Dyeing, and Antibacterial Properties. J. Ind. Eng. Chem. 2025;145:732–744. doi: 10.1016/j.jiec.2024.10.069. [DOI] [Google Scholar]
  175. Kumar P. V., Pushpavalli K. S., Kumar B. G., Kala S. M. J., Prakash K. S.. Green Synthesis of Au-Pt Bimetallic Nanoparticels Using Croton Caudatus Geisel Leaf Extract and Their Biological Studies. Results Chem. 2025;13:101945. doi: 10.1016/j.rechem.2024.101945. [DOI] [Google Scholar]
  176. Sathiyaraj M., Elumalai D., Rajendran V., Hemavathi M., Ashok K., Babu M., Monisha U., Poonguzhali S., Punithavalli S., Abirami G.. Biosynthesis, Characterization, and Multifaceted Applications of Elytraria Acaulis Synthesized Silver and Gold Nanoparticles: Anticancer, Antibacterial, Larvicidal, and Photocatalytic Activities. J. Photochem. Photobiol. B. 2025;263:113102. doi: 10.1016/j.jphotobiol.2025.113102. [DOI] [PubMed] [Google Scholar]
  177. Ali F., Hamza M., Iqbal M., Basha B., Alwadai N., Nazir A.. State-of-Art of Silver and Gold Nanoparticles Synthesis Routes, Characterization and Applications: A Review. Z. Phys. Chem. 2022;236:291–326. doi: 10.1515/zpch-2021-3084. [DOI] [Google Scholar]
  178. Baig N., Kammakakam I., Falath W., Kammakakam I.. Nanomaterials: A Review of Synthesis Methods, Properties, Recent Progress, and Challenges. Mater. Adv. 2021;2:1821–1871. doi: 10.1039/d0ma00807a. [DOI] [Google Scholar]
  179. Al-Harbi N., Abd-Elrahman N. K.. Physical Methods for Preparation of Nanomaterials, Their Characterization and Applications: A Review. J. Umm Al-Qura Univ. Appl. Sci. 2024;11:356–377. doi: 10.1007/s43994-024-00165-7. [DOI] [Google Scholar]
  180. Nguyen M. T., Yonezawa T.. Sputtering onto a Liquid: Interesting Physical Preparation Method for Multi-Metallic Nanoparticles. Sci. Technol. Adv. Mater. 2018;19:883–898. doi: 10.1080/14686996.2018.1542926. [DOI] [Google Scholar]
  181. Patil T., Gambhir R., Vibhute A., Tiwari A. P.. Gold Nanoparticles: Synthesis Methods, Functionalization and Biological Applications. J. Cluster Sci. 2023;34:705–725. doi: 10.1007/s10876-022-02287-6. [DOI] [Google Scholar]
  182. Kareem T. A., Kaliani A. A.. Glow Discharge Plasma Electrolysis for Nanoparticles Synthesis. Ionics. 2012;18:315–327. doi: 10.1007/s11581-011-0639-y. [DOI] [Google Scholar]
  183. De Freitas L. F., Varca G. H. C., Batista J. G. D. S., Lugão A. B.. An Overview of the Synthesis of Gold Nanoparticles Using Radiation Technologies. Nanomaterials. 2018;8:939. doi: 10.3390/nano8110939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Semaltianos N. G.. Nanoparticles by Laser Ablation. Crit. Rev. Solid State Mater. Sci. 2010;35(2):105–124. doi: 10.1080/10408431003788233. [DOI] [Google Scholar]
  185. Hammami I., Alabdallah N. M., jomaa A., Al, kamoun M.. Gold Nanoparticles: Synthesis Properties and Applications. J. King Saud Univ. Sci. 2021;33(7):101560. doi: 10.1016/j.jksus.2021.101560. [DOI] [Google Scholar]
  186. Yeh Y.-C., Creran B., Rotello V. M.. Gold Nanoparticles: Preparation, Properties, and Applications in Bionanotechnology. Nanoscale. 2012;4(6):1871–1880. doi: 10.1039/C1NR11188D. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Herizchi R., Abbasi E., Milani M., Akbarzadeh A.. Current Methods for Synthesis of Gold Nanoparticles. Artif. Cells Nanomed. Biotechnol. 2016;44(2):596–602. doi: 10.3109/21691401.2014.971807. [DOI] [PubMed] [Google Scholar]
  188. Duman H., Akdaşçi E., Eker F., Bechelany M., Karav S.. Gold Nanoparticles: Multifunctional Properties, Synthesis, and Future Prospects. Nanomaterials. 2024;14:1805. doi: 10.3390/nano14221805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Dykman L. A., Khlebtsov N. G.. Methods for Chemical Synthesis of Colloidal Gold. Russ. Chem. Rev. 2019;88(3):229–247. doi: 10.1070/RCR4843. [DOI] [Google Scholar]
  190. Setia, A. ; Mehata, A. K. ; Malik, A. K. ; Muthu, M. S. . Synthesis of Gold Nanoparticles Using Physical and Chemical Methods: Strategies for Stabilization, Controlling Shape, Size and Morphology. In Gold Nanoparticles, Nanomaterials and Nanocomposites; Pasha, S. K. K. ; Deshmukh, K. ; Mustansar Hussain, C. , Eds.; Elsevier, 2025; Chapter 2, pp 41–79 10.1016/B978-0-443-15897-1.00016-9. [DOI] [Google Scholar]
  191. Jamil I., Ahmad F., Khan M. I., Shanableh A., Farooq N., Anjum S., Taj M. B.. A Review of the Gold Nanoparticles’ Synthesis and Application in Dye Degradation. Cleaner Chem. Eng. 2024;10:100126. doi: 10.1016/j.clce.2024.100126. [DOI] [Google Scholar]
  192. Vinnacombe-Willson G. A., Conti Y., Stefancu A., Weiss P. S., Cortés E., Scarabelli L.. Direct Bottom-Up In Situ Growth: A Paradigm Shift for Studies in Wet-Chemical Synthesis of Gold Nanoparticles. Chem. Rev. 2023;123(13):8488–8529. doi: 10.1021/acs.chemrev.2c00914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Hammami I., Alabdallah N. M., Al jomaa A., kamoun M.. Gold Nanoparticles: Synthesis Properties and Applications. J. King Saud Univ. Sci. 2021;33(7):101560. doi: 10.1016/j.jksus.2021.101560. [DOI] [Google Scholar]
  194. Tyas K. P., Maratussolihah P., Rahmadianti S., Girsang G. C. S., Nandiyanto A. B. D.. Review: Comparison of Gold Nanoparticle (AuNP) Synthesis Method. Arabian J. Chem. Environ. Res. 2021;08:436–454. [Google Scholar]
  195. Muddapur U. M., Alshehri S., Ghoneim M. M., Mahnashi M. H., Alshahrani M. A., Khan A. A., Shakeel Iqubal S. M., Bahafi A., More S. S., Shaikh I. A., Mannasaheb B. A., Othman N., Maqbul M. S., Ahmad M. Z.. Plant-Based Synthesis of Gold Nanoparticles and Theranostic Applications: A Review. Molecules. 2022;27:1391. doi: 10.3390/molecules27041391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Santhosh P. B., Genova J., Chamati H.. Review Green Synthesis of Gold Nanoparticles: An Eco-Friendly Approach. Chemistry. 2022;4:345–369. doi: 10.3390/chemistry4020026. [DOI] [Google Scholar]
  197. Perveen F., Ullah F., Rehman A., Zaman H., Abbas W., Khan M., Bilal M., Khan A.. A review on nanotechnology-driven green synthesis of silver nanoparticles using nigella sativa. Insights-J. Health Rehabil. 2025;3(3):121–128. doi: 10.71000/7k52p930. [DOI] [Google Scholar]
  198. Vanga S., Satla S. R.. A Review on Green Synthesis, Characterization and Applications of Plant Mediated Metal Nanoparticles. Next Res. 2025;2(2):100356. doi: 10.1016/j.nexres.2025.100356. [DOI] [Google Scholar]
  199. Bhattarai B., Zaker Y., Bigioni T. P.. Green Synthesis of Gold and Silver Nanoparticles: Challenges and Opportunities. Curr. Opin. Green Sustainable Chem. 2018;12:91–100. doi: 10.1016/j.cogsc.2018.06.007. [DOI] [Google Scholar]
  200. Kirubakaran, D. ; Wahid, J. B. A. ; Karmegam, N. ; Jeevika, R. ; Sellapillai, L. ; Rajkumar, M. ; SenthilKumar, K. J. . A Comprehensive Review on the Green Synthesis of Nanoparticles: Advancements in Biomedical and Environmental Applications Biomed. Mater. Devices 2025. 10.1007/s44174-025-00295-4. [DOI]
  201. Deka K., Nongbet R. D., Das K., Saikia P., Kaur S., Talukder A., Thakuria B.. Understanding the Mechanism Underlying the Green Synthesis of Metallic Nanoparticles Using Plant Extract(s) with Special Reference to Silver, Gold, Copper and Zinc Oxide Nanoparticles. Hybrid Adv. 2025;9:100399. doi: 10.1016/j.hybadv.2025.100399. [DOI] [Google Scholar]
  202. Shamim; Ali, S. ; Ali, T. ; Sharma, H. ; Kishor, B. N. ; Jha, S. K. . Recent Advances in Monodisperse Gold Nanoparticle Delivery, Synthesis, and Emerging Applications in Cancer Therapy Plasmonics 2025. 10.1007/s11468-024-02732-4. [DOI]
  203. Nagpal M., Mittal A., Vinod. Application of Biosurfactants in the Green Synthesis of Inorganic Nanoparticles. Bionanoscience. 2025;15(1):1. doi: 10.1007/s12668-024-01628-1. [DOI] [Google Scholar]
  204. Manyuan N., Danwanichakul P.. Coating Gold Nanoparticles Synthesized from Spent Coffee Ground Extract to Enhance Their Stability. Eng. J. 2021;25(9):55–66. doi: 10.4186/ej.2021.25.9.55. [DOI] [Google Scholar]
  205. Huang X., Devi S., Bordiga M., Brennan C. S., Xu B.. Phenolic Compounds Mediated Biosynthesis of Gold Nanoparticles and Evaluation of Their Bioactivities: A Review. Int. J. Food Sci. Technol. 2023;58:1673–1694. doi: 10.1111/ijfs.16346. [DOI] [Google Scholar]
  206. Santhosh P. B., Genova J., Chamati H.. Review Green Synthesis of Gold Nanoparticles: An Eco-Friendly Approach. Chemistry. 2022;4:345–369. doi: 10.3390/chemistry4020026. [DOI] [Google Scholar]
  207. Rai A., Seena S., Gagliardi T., Palma P. J.. Advances in the Design of Amino Acid and Peptide Synthesized Gold Nanoparticles for Their Applications. Adv. Colloid Interface Sci. 2023;318:102951. doi: 10.1016/j.cis.2023.102951. [DOI] [PubMed] [Google Scholar]
  208. Gholami-Shabani M., Shams-Ghahfarokhi M., Gholami-Shabani Z., Akbarzadeh A., Riazi G., Ajdari S., Amani A., Razzaghi-Abyaneh M.. Enzymatic Synthesis of Gold Nanoparticles Using Sulfite Reductase Purified from Escherichia Coli: A Green Eco-Friendly Approach. Process Biochem. 2015;50(7):1076–1085. doi: 10.1016/j.procbio.2015.04.004. [DOI] [Google Scholar]
  209. Shedbalkar U., Singh R., Wadhwani S., Gaidhani S., Chopade B. A.. Microbial Synthesis of Gold Nanoparticles: Current Status and Future Prospects. Adv. Colloid Interface Sci. 2014;209:40–48. doi: 10.1016/j.cis.2013.12.011. [DOI] [PubMed] [Google Scholar]
  210. Pattnayak, S. ; Choudhury, S. ; Hota, G. . Green Chemistry Mediated and Biogenic Synthesis of Gold Nanoparticles: Prospects and Challenges. In Gold Nanoparticles, Nanomaterials and Nanocomposites: Science, Technology and Applications; Elsevier, 2025; pp 81–111 10.1016/B978-0-443-15897-1.00003-0. [DOI] [Google Scholar]
  211. Verma J., Kumar C., Sharma M., Saxena S.. Biotechnological Advances in Microbial Synthesis of Gold Nanoparticles: Optimizations and Applications. 3 Biotech. 2024;14:263. doi: 10.1007/s13205-024-04110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Stozhko N. Y., Bukharinova M. A., Khamzina E. I., Tarasov A. V., Vidrevich M. B., Brainina K. Z.. The Effect of the Antioxidant Activity of Plant Extracts on the Properties of Gold Nanoparticles. Nanomaterials. 2019;9(12):1655. doi: 10.3390/nano9121655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Elia P., Zach R., Hazan S., Kolusheva S., Porat Z., Zeiri Y.. Green Synthesis of Gold Nanoparticles Using Plant Extracts as Reducing Agents. Int. J. Nanomedicine. 2014;9(1):4007–4021. doi: 10.2147/IJN.S57343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Fanoro O. T., Oluwafemi O. S.. Bactericidal Antibacterial Mechanism of Plant Synthesized Silver, Gold and Bimetallic Nanoparticles. Pharmaceutics. 2020;12:1044. doi: 10.3390/pharmaceutics12111044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Hassanisaadi M., Bonjar G. H. S., Rahdar A., Pandey S., Hosseinipour A., Abdolshahi R.. Environmentally Safe Biosynthesis of Gold Nanoparticles Using Plant Water Extracts. Nanomaterials. 2021;11(8):2033. doi: 10.3390/nano11082033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Siddiqi K. S., Husen A.. Engineered Gold Nanoparticles and Plant Adaptation Potential. Nanoscale Res. Lett. 2016;11:400. doi: 10.1186/s11671-016-1607-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Timoszyk A., Grochowalska R.. Mechanism and Antibacterial Activity of Gold Nanoparticles (AuNPs) Functionalized with Natural Compounds from Plants. Pharmaceutics. 2022;14:2599. doi: 10.3390/pharmaceutics14122599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Paul B., Bhuyan B., Purkayastha D. D., Dhar S. S.. Photocatalytic and Antibacterial Activities of Gold and Silver Nanoparticles Synthesized Using Biomass of Parkia Roxburghii Leaf. J. Photochem. Photobiol. B. 2016;154:1–7. doi: 10.1016/j.jphotobiol.2015.11.004. [DOI] [PubMed] [Google Scholar]
  219. Philip D., Unni C., Aromal S. A., Vidhu V. K.. Murraya Koenigii Leaf-Assisted Rapid Green Synthesis of Silver and Gold Nanoparticles. Spectrochim. Acta, Part A. 2011;78(2):899–904. doi: 10.1016/j.saa.2010.12.060. [DOI] [PubMed] [Google Scholar]
  220. Rauf A., Ahmad T., Khan A., Maryam, Uddin G., Ahmad B., Mabkhot Y. N., Bawazeer S., Riaz N., Malikovna B. K., Almarhoon Z. M., Al-Harrasi A.. Green Synthesis and Biomedicinal Applications of Silver and Gold Nanoparticles Functionalized with Methanolic Extract of Mentha Longifolia. Artif. Cells Nanomed. Biotechnol. 2021;49(1):194–203. doi: 10.1080/21691401.2021.1890099. [DOI] [PubMed] [Google Scholar]
  221. Sheny D. S., Mathew J., Philip D.. Phytosynthesis of Au, Ag and Au–Ag Bimetallic Nanoparticles Using Aqueous Extract and Dried Leaf of Anacardium Occidentale. Spectrochim. Acta, Part A. 2011;79(1):254–262. doi: 10.1016/j.saa.2011.02.051. [DOI] [PubMed] [Google Scholar]
  222. Singh P., Ahn S., Kang J. P., Veronika S., Huo Y., Singh H., Chokkaligam M., El-Agamy Farh M., Aceituno V. C., Kim Y. J., Yang D. C.. In Vitro Anti-Inflammatory Activity of Spherical Silver Nanoparticles and Monodisperse Hexagonal Gold Nanoparticles by Fruit Extract of Prunus Serrulata: A Green Synthetic Approach. Artif. Cells Nanomed. Biotechnol. 2018;46(8):2022–2032. doi: 10.1080/21691401.2017.1408117. [DOI] [PubMed] [Google Scholar]
  223. Velmurugan P., Anbalagan K., Manosathyadevan M., Lee K. J., Cho M., Lee S. M., Park J. H., Oh S. G., Bang K. S., Oh B. T.. Green Synthesis of Silver and Gold Nanoparticles Using Zingiber Officinale Root Extract and Antibacterial Activity of Silver Nanoparticles against Food Pathogens. Bioprocess Biosyst. Eng. 2014;37(10):1935–1943. doi: 10.1007/s00449-014-1169-6. [DOI] [PubMed] [Google Scholar]
  224. Vijayan R., Joseph S., Mathew B.. Indigofera Tinctoria Leaf Extract Mediated Green Synthesis of Silver and Gold Nanoparticles and Assessment of Their Anticancer, Antimicrobial, Antioxidant and Catalytic Properties. Artif. Cells Nanomed. Biotechnol. 2018;46(4):861–871. doi: 10.1080/21691401.2017.1345930. [DOI] [PubMed] [Google Scholar]
  225. Vijayan R., Joseph S., Mathew B.. Anticancer, Antimicrobial, Antioxidant, and Catalytic Activities of Green-Synthesized Silver and Gold Nanoparticles Using Bauhinia Purpurea Leaf Extract. Bioprocess Biosyst. Eng. 2019;42(2):305–319. doi: 10.1007/s00449-018-2035-8. [DOI] [PubMed] [Google Scholar]
  226. Wang D., Markus J., Wang C., Kim Y. J., Mathiyalagan R., Aceituno V. C., Ahn S., Yang D. C.. Green Synthesis of Gold and Silver Nanoparticles Using Aqueous Extract of Cibotium Barometz Root. Artif. Cells Nanomed. Biotechnol. 2017;45(8):1548–1555. doi: 10.1080/21691401.2016.1260580. [DOI] [PubMed] [Google Scholar]
  227. Yallappa S., Manjanna J., Dhananjaya B. L.. Phytosynthesis of Stable Au, Ag and Au–Ag Alloy Nanoparticles Using J. Sambac Leaves Extract, and Their Enhanced Antimicrobial Activity in Presence of Organic Antimicrobials. Spectrochim. Acta, Part A. 2015;137(1):236–243. doi: 10.1016/j.saa.2014.08.030. [DOI] [PubMed] [Google Scholar]
  228. Dhayalan M., Denison M. I. J., Ayyar M., Gandhi N. N., Krishnan K., Abdulhadi B.. Biogenic Synthesis, Characterization of Gold and Silver Nanoparticles from Coleus Forskohlii and Their Clinical Importance. J. Photochem. Photobiol. B. 2018;183:251–257. doi: 10.1016/j.jphotobiol.2018.04.042. [DOI] [PubMed] [Google Scholar]
  229. Naraginti S., Sivakumar A.. Eco-Friendly Synthesis of Silver and Gold Nanoparticles with Enhanced Bactericidal Activity and Study of Silver Catalyzed Reduction of 4-Nitrophenol. Spectrochim. Acta, Part A. 2014;128:357–362. doi: 10.1016/j.saa.2014.02.083. [DOI] [PubMed] [Google Scholar]
  230. Elavazhagan T., Arunachalam K. D.. Memecylon Edule Leaf Extract Mediated Green Synthesis of Silver and Gold Nanoparticles. Int. J. Nanomed. 2011;6:1265–1278. doi: 10.2147/ijn.s18347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Francis S., Joseph S., Koshy E. P., Mathew B.. Green Synthesis and Characterization of Gold and Silver Nanoparticles Using Mussaenda Glabrata Leaf Extract and Their Environmental Applications to Dye Degradation. Environ. Sci. Pollut. Res. 2017;24(21):17347–17357. doi: 10.1007/s11356-017-9329-2. [DOI] [PubMed] [Google Scholar]
  232. Francis S., Koshy E. P., Mathew B.. Green Synthesis of Stereospermum Suaveolens Capped Silver and Gold Nanoparticles and Assessment of Their Innate Antioxidant, Antimicrobial and Antiproliferative Activities. Bioprocess Biosyst. Eng. 2018;41(7):939–951. doi: 10.1007/s00449-018-1925-0. [DOI] [PubMed] [Google Scholar]
  233. Godipurge S. S., Yallappa S., Biradar N. J., Biradar J. S., Dhananjaya B. L., Hegde G., Jagadish K., Hegde G.. A Facile and Green Strategy for the Synthesis of Au, Ag and Au–Ag Alloy Nanoparticles Using Aerial Parts of R. Hypocrateriformis Extract and Their Biological Evaluation. Enzyme Microb. Technol. 2016;95:174–184. doi: 10.1016/j.enzmictec.2016.08.006. [DOI] [PubMed] [Google Scholar]
  234. Gopinath K., Kumaraguru S., Bhakyaraj K., Mohan S., Venkatesh K. S., Esakkirajan M., Kaleeswarran P., Alharbi N. S., Kadaikunnan S., Govindarajan M., Benelli G., Arumugam A.. Green Synthesis of Silver, Gold and Silver/Gold Bimetallic Nanoparticles Using the Gloriosa Superba Leaf Extract and Their Antibacterial and Antibiofilm Activities. Microb. Pathog. 2016;101:1–11. doi: 10.1016/j.micpath.2016.10.011. [DOI] [PubMed] [Google Scholar]
  235. Huo Y., Singh P., Kim Y. J., Soshnikova V., Kang J., Markus J., Ahn S., Castro-Aceituno V., Mathiyalagan R., Chokkalingam M., Bae K. S., Yang D. C.. Biological Synthesis of Gold and Silver Chloride Nanoparticles by Glycyrrhiza Uralensis and in Vitro Applications. Artif. Cells Nanomed. Biotechnol. 2018;46(2):303–312. doi: 10.1080/21691401.2017.1307213. [DOI] [PubMed] [Google Scholar]
  236. Joseph S., Mathew B.. Microwave Assisted Facile Green Synthesis of Silver and Gold Nanocatalysts Using the Leaf Extract of Aerva Lanata. Spectrochim. Acta, Part A. 2015;136(PC):1371–1379. doi: 10.1016/j.saa.2014.10.023. [DOI] [PubMed] [Google Scholar]
  237. Wang C., Mathiyalagan R., Kim Y. J., Castro-Aceituno V., Singh P., Ahn S., Wang D., Yang D. C.. Rapid Green Synthesis of Silver and Gold Nanoparticles Using Dendropanax Morbifera Leaf Extract and Their Anticancer Activities. Int. J. Nanomed. 2016;11:3691–3701. doi: 10.2147/IJN.S97181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Markus J., Wang D., Kim Y. J., Ahn S., Mathiyalagan R., Wang C., Yang D. C.. Biosynthesis, Characterization, and Bioactivities Evaluation of Silver and Gold Nanoparticles Mediated by the Roots of Chinese Herbal Angelica Pubescens Maxim. Nanoscale Res. Lett. 2017;12(1):46. doi: 10.1186/s11671-017-1833-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Nayem S. M. A., Sultana N., Haque M. A., Miah B., Hasan M. M., Islam T., Hasan M. M., Awal A., Uddin J., Aziz M. A., Ahammad A. J. S.. Green Synthesis of Gold and Silver Nanoparticles by Using Amorphophallus Paeoniifolius Tuber Extract and Evaluation of Their Antibacterial Activity. Molecules. 2020;25(20):4773. doi: 10.3390/molecules25204773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Amina M., Al Musayeib N. M., Alarfaj N. A., El-Tohamy M. F., Al-Hamoud G. A.. Antibacterial and Immunomodulatory Potentials of Biosynthesized Ag, Au, Ag-Au Bimetallic Alloy Nanoparticles Using the Asparagus Racemosus Root Extract. Nanomaterials. 2020;10(12):2453. doi: 10.3390/nano10122453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Arunachalam K. D., Annamalai S. K.. Chrysopogon Zizanioides Aqueous Extract Mediated Synthesis, Characterization of Crystalline Silver and Gold Nanoparticles for Biomedical Applications. Int. J. Nanomed. 2013;8:2375–2384. doi: 10.2147/IJN.S44076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Arunachalam K. D., Annamalai S. K., Hari S.. One-Step Green Synthesis and Characterization of Leaf Extract-Mediated Biocompatible Silver and Gold Nanoparticles from Memecylon Umbellatum. Int. J. Nanomed. 2013;8:1307–1315. doi: 10.2147/IJN.S36670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Khan M., Al-Hamoud K., Liaqat Z., Shaik M. R., Adil S. F., Kuniyil M., Alkhathlan H. Z., Al-Warthan A., Siddiqui M. R. H., Mondeshki M., Tremel W., Khan M., Tahir M. N.. Synthesis of Au, Ag, and Au–Ag Bimetallic Nanoparticles Using Pulicaria Undulata Extract and Their Catalytic Activity for the Reduction of 4-Nitrophenol. Nanomaterials. 2020;10(9):1885. doi: 10.3390/nano10091885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Singh P., Pandit S., Beshay M., Mokkapati V. R. S. S., Garnaes J., Olsson M. E., Sultan A., Mackevica A., Mateiu R. V., Lütken H., Daugaard A. E., Baun A., Mijakovic I.. Anti-Biofilm Effects of Gold and Silver Nanoparticles Synthesized by the Rhodiola Rosea Rhizome Extracts. Artif. Cells Nanomed. Biotechnol. 2018;46(sup3):886–899. doi: 10.1080/21691401.2018.1518909. [DOI] [PubMed] [Google Scholar]
  245. Park J. S., Ahn E. Y., Park Y.. Asymmetric Dumbbell-Shaped Silver Nanoparticles and Spherical Gold Nanoparticles Green-Synthesized by Mangosteen (Garcinia Mangostana) Pericarp Waste Extracts. Int. J. Nanomed. 2017;12:6895–6908. doi: 10.2147/IJN.S140190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Bonigala B., Kasukurthi B., Konduri V. V., Mangamuri U. K., Gorrepati R., Poda S.. Green Synthesis of Silver and Gold Nanoparticles Using Stemona Tuberosa Lour and Screening for Their Catalytic Activity in the Degradation of Toxic Chemicals. Environ. Sci. Pollut. Res. 2018;25(32):32540–32548. doi: 10.1007/s11356-018-3105-9. [DOI] [PubMed] [Google Scholar]
  247. Naraginti S., Li Y.. Preliminary Investigation of Catalytic, Antioxidant, Anticancer and Bactericidal Activity of Green Synthesized Silver and Gold Nanoparticles Using Actinidia Deliciosa. J. Photochem. Photobiol. B. 2017;170:225–234. doi: 10.1016/j.jphotobiol.2017.03.023. [DOI] [PubMed] [Google Scholar]
  248. Cardoso-Avila P. E., Patakfalvi R., Rodríguez-Pedroza C., Aparicio-Fernández X., Loza-Cornejo S., Villa-Cruz V., Martínez-Cano E.. One-Pot Green Synthesis of Gold and Silver Nanoparticles Using: Rosa Canina L. Extract. RSC Adv. 2021;11(24):14624–14631. doi: 10.1039/D1RA01448J. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Akinfenwa A. O., Abdul N. S., Docrat F. T., Marnewick J. L., Luckay R. C., Hussein A. A.. Cytotoxic Effects of Phytomediated Silver and Gold Nanoparticles Synthesised from Rooibos (Aspalathus linearis), and Aspalathin. Plants. 2021;10(11):2460. doi: 10.3390/plants10112460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Zayed M. F., Eisa W. H., El-kousy S. M., Mleha W. K., Kamal N.. Ficus Retusa-Stabilized Gold and Silver Nanoparticles: Controlled Synthesis, Spectroscopic Characterization, and Sensing Properties. Spectrochim. Acta, Part A. 2019;214:496–512. doi: 10.1016/j.saa.2019.02.042. [DOI] [PubMed] [Google Scholar]
  251. Devi G. K., Sathishkumar K.. Synthesis of Gold and Silver Nanoparticles Using Mukia Maderaspatna Plant Extract and Its Anticancer Activity. IET Nanobiotechnol. 2017;11(2):143–151. doi: 10.1049/iet-nbt.2015.0054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Khan S. A., Shahid S., Lee C. S.. Green Synthesis of Gold and Silver Nanoparticles Using Leaf Extract of Clerodendrum Inerme; Characterization, Antimicrobial, and Antioxidant Activities. Biomolecules. 2020;10(6):835. doi: 10.3390/biom10060835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Ahmad N., Sharma A. K., Sharma S., Khan I., Sharma D. K., Shamsi A., Kumar T. R. S., Seervi M.. Biosynthesized Composites of Au-Ag Nanoparticles Using Trapa Peel Extract Induced ROS-Mediated P53 Independent Apoptosis in Cancer Cells. Drug Chem. Toxicol. 2019;42(1):43–53. doi: 10.1080/01480545.2018.1463241. [DOI] [PubMed] [Google Scholar]
  254. Pérez Z. E. J., Mathiyalagan R., Markus J., Kim Y. J., Kang H. M., Abbai R., Seo K. H., Wang D., Soshnikova V., Yang D. C.. Ginseng-Berry-Mediated Gold and Silver Nanoparticle Synthesis and Evaluation of Their in Vitro Antioxidant, Antimicrobial, and Cytotoxicity Effects on Human Dermal Fibroblast and Murine Melanoma Skin Cell Lines. Int. J. Nanomed. 2017;12:709–723. doi: 10.2147/IJN.S118373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Abbai R., Mathiyalagan R., Markus J., Kim Y. J., Wang C., Singh P., Ahn S., Farh M. E. A., Yang D. C.. Green Synthesis of Multifunctional Silver and Gold Nanoparticles from the Oriental Herbal Adaptogen: Siberian Ginseng. Int. J. Nanomed. 2016;11:3131–3143. doi: 10.2147/IJN.S108549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Filip G. A., Moldovan B., Baldea I., Olteanu D., Suharoschi R., Decea N., Cismaru C. M., Gal E., Cenariu M., Clichici S., David L.. UV-Light Mediated Green Synthesis of Silver and Gold Nanoparticles Using Cornelian Cherry Fruit Extract and Their Comparative Effects in Experimental Inflammation. J. Photochem. Photobiol. B. 2019;191:26–37. doi: 10.1016/j.jphotobiol.2018.12.006. [DOI] [PubMed] [Google Scholar]
  257. Zuhrotun A., Oktaviani D. J., Hasanah A. N.. Biosynthesis of Gold and Silver Nanoparticles Using Phytochemical Compounds. Molecules. 2023;27:3240. doi: 10.3390/molecules28073240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Chaurasia P. K., Bharati S. L., Yadava S.. Nano-Reduction of Gold and Silver Ions: A Perspective on the Fate of Microbial Laccases as Potential Biocatalysts in the Synthesis of Metals (Gold and Silver) Nano-Particles. Curr. Res. Microb. Sci. 2022;3:100098. doi: 10.1016/j.crmicr.2021.100098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Duman H., Akdaşçi E., Eker F., Bechelany M., Karav S.. Gold Nanoparticles: Multifunctional Properties, Synthesis, and Future Prospects. Nanomaterials. 2024;14:1805. doi: 10.3390/nano14221805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Li M., Cushing S. K., Wu N.. Plasmon-Enhanced Optical Sensors: A Review. Analyst. 2015;140(2):386–406. doi: 10.1039/C4AN01079E. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Yeh Y.-C., Creran B., Rotello V. M.. Gold Nanoparticles: Preparation, Properties, and Applications in Bionanotechnology. Nanoscale. 2012;4(6):1871–1880. doi: 10.1039/C1NR11188D. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Yue J., Feliciano T. J., Li W., Lee A., Odom T. W.. Gold Nanoparticle Size and Shape Effects on Cellular Uptake and Intracellular Distribution of SiRNA Nanoconstructs. Bioconjugate Chem. 2017;28(6):1791–1800. doi: 10.1021/acs.bioconjchem.7b00252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Xie X., Liao J., Shao X., Li Q., Lin Y.. The Effect of Shape on Cellular Uptake of Gold Nanoparticles in the Forms of Stars, Rods, and Triangles. Sci. Rep. 2017;7(1):3827. doi: 10.1038/s41598-017-04229-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Sani A., Cao C., Cui D.. Toxicity of Gold Nanoparticles (AuNPs): A Review. Biochem. Biophys. Rep. 2021;26:100991. doi: 10.1016/j.bbrep.2021.100991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Seitz O., Chehimi M. M., Cabet-Deliry E., Truong S., Felidj N., Perruchot C., Greaves S. J., Watts J. F.. Preparation and Characterisation of Gold Nanoparticle Assemblies on Silanised Glass Plates. Colloids Surf., A. 2003;218(1):225–239. doi: 10.1016/S0927-7757(02)00594-0. [DOI] [Google Scholar]
  266. Rasheed, S. A. ; Nayef, U. M. . Effect of Laser Energy on Formation of Gold@Aluminum Oxide (Core@Shell) Nanoparticles for Enhancement Spectral Responsivity Plasmonics 2025. 10.1007/s11468-025-02842-7. [DOI]
  267. Amendola V., Meneghetti M.. Size Evaluation of Gold Nanoparticles by UV–vis Spectroscopy. J. Phys. Chem. C. 2009;113(11):4277–4285. doi: 10.1021/jp8082425. [DOI] [Google Scholar]
  268. Wang L., Wei Y., Liu X., Chen J., Mao C., Jin B.. Electrochemiluminescence Biosensor Based on Gold Nanoparticles Modulated Cathode AIE-Activated Metal-Organic Frameworks for the Ultrasensitive Detection of CA15–3. Biosens. Bioelectron. 2025;282:117465. doi: 10.1016/j.bios.2025.117465. [DOI] [PubMed] [Google Scholar]
  269. Iqbal Y., Amin F., Aziz M. H., Alhadlaq H. A., Alaizeri Z. M.. Biomimetic Sodium Alginate Functionalized Gold Nanoparticles Loaded with Trans-Resveratrol for in-Vitro Cancer Treatment and Intercellular ROS Studies against MCF-7 Cell Lines. Colloids Surf., A. 2025;712:136448. doi: 10.1016/j.colsurfa.2025.136448. [DOI] [Google Scholar]
  270. Danchova N., Tsekov R., Shandurkov D., Gutzov S., Lyubenova L., Mihaylov L., Spassov T.. Silica Gels Doped with Gold Nanoparticles and Gold Thiolate Complexes: The Effect of Heating and Preparation Conditions. J. Non Cryst. Solids. 2025;648:123308. doi: 10.1016/j.jnoncrysol.2024.123308. [DOI] [Google Scholar]
  271. Ahmady I. M., Parambath J. B. M., Elsheikh E. A. E., Kim G., Han C., Pérez-García A., Mohamed A. A.. Bacterial Synthesis of Anisotropic Gold Nanoparticles. Appl. Microbiol. Biotechnol. 2025;109(1):62. doi: 10.1007/s00253-025-13438-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Duman H., Akdaşçi E., Eker F., Bechelany M., Karav S.. Gold Nanoparticles: Multifunctional Properties, Synthesis, and Future Prospects. Nanomaterials. 2024;14(22):1805. doi: 10.3390/nano14221805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Wang L., Wei Y., Liu X., Chen J., Mao C., Jin B.. Electrochemiluminescence Biosensor Based on Gold Nanoparticles Modulated Cathode AIE-Activated Metal-Organic Frameworks for the Ultrasensitive Detection of CA15–3. Biosens. Bioelectron. 2025;282:117465. doi: 10.1016/j.bios.2025.117465. [DOI] [PubMed] [Google Scholar]
  274. Oliveira A. E. F., Pereira A. C., Resende M. A. C., Ferreira L. F.. Gold Nanoparticles: A Didactic Step-by-Step of the Synthesis Using the Turkevich Method, Mechanisms, and Characterizations. Analytica. 2023;4(2):250–263. doi: 10.3390/analytica4020020. [DOI] [Google Scholar]
  275. Mehra V., Kumar S., Tamang A. M., Chandraker S. K.. Green Synthesis of Gold Nanoparticles (AuNPs) by Using Plant Extract and Their Biological Application: A Review. Bionanoscience. 2025;15(1):18. doi: 10.1007/s12668-024-01703-7. [DOI] [Google Scholar]
  276. Ngo V. K. T., Nguyen D. G., Huynh T. P., Lam Q. V.. A Low Cost Technique for Synthesis of Gold Nanoparticles Using Microwave Heating and Its Application in Signal Amplification for Detecting Escherichia Coli O157:H7 Bacteria. Adv. Nat. Sci.: Nanosci. Nanotechnol. 2016;7(3):035016. doi: 10.1088/2043-6262/7/3/035016. [DOI] [Google Scholar]
  277. Eid, M. M. Characterization of Nanoparticles by FTIR and FTIR-Microscopy. In Handbook of Consumer Nanoproducts; Springer: Singapore, 2021; pp 1–30 10.1007/978-981-15-6453-6_89-1. [DOI] [Google Scholar]
  278. Hutchinson N., Wu Y., Wang Y., Kanungo M., Debruine A., Kroll E., Gilmore D., Eckrose Z., Gaston S., Matel P., Kaltchev M., Nickel A. M., Kumpaty S., Hua X., Zhang W.. Green Synthesis of Gold Nanoparticles Using Upland Cress and Their Biochemical Characterization and Assessment. Nanomaterials. 2022;12(1):28. doi: 10.3390/nano12010028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Alhiti L. S., Hashim E. H., Abdullah F. H.. Green Methods for Gold Nanoparticle Synthesis: Properties, Characterization, and Diverse Applications – Review Article. Humanitarian Nat. Sci. J. 2025;6(2):249–265. doi: 10.53796/hnsj63/12. [DOI] [Google Scholar]
  280. Seitz O., Chehimi M. M., Cabet-Deliry E., Truong S., Felidj N., Perruchot C., Greaves S. J., Watts J. F.. Preparation and Characterisation of Gold Nanoparticle Assemblies on Silanised Glass Plates. Colloids Surf., A. 2003;218(1):225–239. doi: 10.1016/S0927-7757(02)00594-0. [DOI] [Google Scholar]
  281. Isa S. Z. M., Zainon R., Tamal M.. State of the Art in Gold Nanoparticle Synthesisation via Pulsed Laser Ablation in Liquid and Its Characterisation for Molecular Imaging: A Review. Materials. 2022;15:875. doi: 10.3390/ma15030875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Koliyote S., Shaji J.. A Recent Review on Synthesis, Characterization and Activities of Gold Nanoparticles Using Plant Extracts. Indian J. Pharm. Educ. Res. 2023;57:s198–s212. doi: 10.5530/ijper.57.2s.24. [DOI] [Google Scholar]
  283. Maseko N. N., Enke D., Owolawi P. A., Iwarere S. A., Oluwafemi O. S., Pocock J.. The Influence of Gold Nanoparticles Addition on Sugarcane Leaves-Derived Silica Xerogel Catalyst for the Production of Biodiesel. Gels. 2025;11(3):153. doi: 10.3390/gels11030153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Iqbal Y., Amin F., Aziz M. H., Alhadlaq H. A., Alaizeri Z. M.. Biomimetic Sodium Alginate Functionalized Gold Nanoparticles Loaded with Trans-Resveratrol for in-Vitro Cancer Treatment and Intercellular ROS Studies against MCF-7 Cell Lines. Colloids Surf., A. 2025;712:136448. doi: 10.1016/j.colsurfa.2025.136448. [DOI] [Google Scholar]
  285. Mehra V., Kumar S., Tamang A. M., Chandraker S. K.. Green Synthesis of Gold Nanoparticles (AuNPs) by Using Plant Extract and Their Biological Application: A Review. Bionanoscience. 2025;15(1):18. doi: 10.1007/s12668-024-01703-7. [DOI] [Google Scholar]
  286. Danchova N., Tsekov R., Shandurkov D., Gutzov S., Lyubenova L., Mihaylov L., Spassov T.. Silica Gels Doped with Gold Nanoparticles and Gold Thiolate Complexes: The Effect of Heating and Preparation Conditions. J. Non Cryst. Solids. 2025;648:123308. doi: 10.1016/j.jnoncrysol.2024.123308. [DOI] [Google Scholar]
  287. Mostafa A. M., Mwafy E. A.. Synthesis of ZnO and Au@ZnO Core/Shell Nano-Catalysts by Pulsed Laser Ablation in Different Liquid Media. J. Mater. Res. Technol. 2020;9(3):3241–3248. doi: 10.1016/j.jmrt.2020.01.071. [DOI] [Google Scholar]
  288. Mostafa A. M., Mwafy E. A.. Effect of Dual-Beam Laser Radiation for Synthetic SnO2/Au Nanoalloy for Antibacterial Activity. J. Mol. Struct. 2020;1222:128913. doi: 10.1016/j.molstruc.2020.128913. [DOI] [Google Scholar]
  289. Yue Y., Moriyama A., Mita M., Yu Y.. Sustainable Synthesis, Characterization, Cellular Effects of Gold Nanoparticles and Their Applications as Therapeutics in Cancer Therapy. ChemPlusChem. 2025;90:e202400783. doi: 10.1002/cplu.202400783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Pulit J., Banach M.. Environment-friendly method for obtaining gold nanoparticles based on plant extracT. Digest J. Nanomater. Biostruct. 2013;8:1295–1300. [Google Scholar]
  291. Bhau B. S., Ghosh S., Puri S., Borah B., Sarmah D. K., Khan R.. Green Synthesis of Gold Nanoparticles from the Leaf Extract of Nepenthes Khasiana and Antimicrobial Assay. Adv. Mater. Lett. 2015;6(1):55–58. doi: 10.5185/amlett.2015.5609. [DOI] [Google Scholar]
  292. Ganesan R. M., Gurumallesh Prabu H.. Synthesis of Gold Nanoparticles Using Herbal Acorus Calamus Rhizome Extract and Coating on Cotton Fabric for Antibacterial and UV Blocking Applications. Arabian J. Chem. 2019;12(8):2166–2174. doi: 10.1016/j.arabjc.2014.12.017. [DOI] [Google Scholar]
  293. Mahoney J., Monroe C., Swartley A. M., Ucak-Astarlioglu M. G., Zoto C. A.. Surface Analysis Using X-Ray Photoelectron Spectroscopy. Spectrosc. Lett. 2020;53(10):726–736. doi: 10.1080/00387010.2020.1824197. [DOI] [Google Scholar]
  294. Ielo I., Rando G., Giacobello F., Sfameni S., Castellano A., Galletta M., Drommi D., Rosace G., Plutino M. R.. Synthesis, Chemical–Physical Characterization, and Biomedical Applications of Functional Gold Nanoparticles: A Review. Molecules. 2021;26(19):5823. doi: 10.3390/molecules26195823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  295. Tendo S., Niozu A., Yoshioka K., Tabuse M., Adachi J., Tanaka H., Wada S.. Comparative Study of Electron Transport through Aromatic Molecules on Gold Nanoparticles: Insights from Soft X-Ray Spectroscopy of Condensed Nanoparticle Films versus Flat Monolayer Films. Phys. Chem. Chem. Phys. 2024;27(1):388–396. doi: 10.1039/D4CP03556A. [DOI] [PubMed] [Google Scholar]
  296. Ahmady I. M., Parambath J. B. M., Elsheikh E. A. E., Kim G., Han C., Pérez-García A., Mohamed A. A.. Bacterial Synthesis of Anisotropic Gold Nanoparticles. Appl. Microbiol. Biotechnol. 2025;109(1):62. doi: 10.1007/s00253-025-13438-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Li Y., Dong Z., Wang X., Wang Y., Fan W.. The Accumulation and Tissue Distribution of Gold Nanoparticles in Exposed Pregnant Rats. Environ. Sci.: Nano. 2025;12:3476–3487. doi: 10.1039/D5EN00164A. [DOI] [Google Scholar]
  298. Nene, A. ; Antarnusa, G. ; Dulta, K. ; Sadeghzade, S. ; Wang, L. ; Ravikumar, C. H. ; Aman, J. ; Khongthaw, B. ; Kandwal, A. ; Somani, P. ; Kumar, A. ; Ramachandran, K. ; Subramaniam, V. ; Galluzzi, M. ; Dou, S. ; Liu, X. . Au–Ag Bimetallic Nanoparticles: Synthesis, Structure, and Application in Sensing ChemPhysMater 2025. 10.1016/j.chphma.2025.02.006. [DOI]
  299. Liu S., Lämmerhofer M.. Functionalized Gold Nanoparticles for Sample Preparation: A Review. Electrophoresis. 2019;40(18–19):2438–2461. doi: 10.1002/elps.201900111. [DOI] [PubMed] [Google Scholar]
  300. Fleming D. A., Williams M. E.. Size-Controlled Synthesis of Gold Nanoparticles via High-Temperature Reduction. Langmuir. 2004;20(8):3021–3023. doi: 10.1021/la0362829. [DOI] [PubMed] [Google Scholar]
  301. Mehra V., Kumar S., Tamang A. M., Chandraker S. K.. Green Synthesis of Gold Nanoparticles (AuNPs) by Using Plant Extract and Their Biological Application: A Review. Bionanoscience. 2025;15(1):18. doi: 10.1007/s12668-024-01703-7. [DOI] [Google Scholar]
  302. Danchova N., Tsekov R., Shandurkov D., Gutzov S., Lyubenova L., Mihaylov L., Spassov T.. Silica Gels Doped with Gold Nanoparticles and Gold Thiolate Complexes: The Effect of Heating and Preparation Conditions. J. Non Cryst. Solids. 2025;648:123308. doi: 10.1016/j.jnoncrysol.2024.123308. [DOI] [Google Scholar]
  303. Ahmady I. M., Parambath J. B. M., Elsheikh E. A. E., Kim G., Han C., Pérez-García A., Mohamed A. A.. Bacterial Synthesis of Anisotropic Gold Nanoparticles. Appl. Microbiol. Biotechnol. 2025;109(1):62. doi: 10.1007/s00253-025-13438-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  304. Liu H., Pierre-Pierre N., Huo Q.. Dynamic Light Scattering for Gold Nanorod Size Characterization and Study of Nanorod–Protein Interactions. Gold Bull. 2012;45(4):187–195. doi: 10.1007/s13404-012-0067-4. [DOI] [Google Scholar]
  305. Mapala K., Pattabi M.. Mimosa Pudica Flower Extract Mediated Green Synthesis of Gold Nanoparticles. Nano World J. 2017;03(02):44–50. doi: 10.17756/nwj.2017-045. [DOI] [Google Scholar]
  306. Yue Y., Moriyama A., Mita M., Yu Y.. Sustainable Synthesis, Characterization, Cellular Effects of Gold Nanoparticles and Their Applications as Therapeutics in Cancer Therapy. ChemPlusChem. 2025;90:e202400783. doi: 10.1002/cplu.202400783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  307. Kumar B., Smita K., Vizuete K. S., Cumbal L.. Aqueous Phase Lavender Leaf Mediated Green Synthesis of Gold Nanoparticles and Evaluation of Its Antioxidant Activity. Biol. Med. 2016;08(3):1. doi: 10.4172/0974-8369.1000290. [DOI] [Google Scholar]
  308. Rajakumar G., Gomathi T., Rahuman A. A., Thiruvengadam M., Mydhili G., Kim S.-H., Lee T.-J., Chung I.-M.. Biosynthesis and Biomedical Applications of Gold Nanoparticles Using Eclipta Prostrata Leaf Extract. Appl. Sci. 2016;6(8):222. doi: 10.3390/app6080222. [DOI] [Google Scholar]
  309. Abbasi T., Anuradha J., Abbasi S. A.. Utilization of the Terrestrial Weed Guduchi (Tinospora Cordifolia) in Clean-Green Synthesis of Gold Nanoparticles. Nanosci. Technol. 2014;1:1–7. [Google Scholar]
  310. Dey A., Yogamoorthy A., Sundarapandian S. M.. Green synthesis of gold nanoparticles and evaluation of its cytotoxic property against colon cancer cell line. Res. J. Life Sci., Bioinf., Pharm. Chem. Sci. 2018:1–17. doi: 10.26479/2018.0406.01. [DOI] [Google Scholar]
  311. Wani I. A., Ahmad T.. Size and Shape Dependant Antifungal Activity of Gold Nanoparticles: A Case Study of Candida. Colloids Surf., B. 2013;101:162–170. doi: 10.1016/j.colsurfb.2012.06.005. [DOI] [PubMed] [Google Scholar]
  312. Koul B., Poonia A. K., Yadav D., Jin J. O.. Microbe-Mediated Biosynthesis of Nanoparticles: Applications and Future Prospects. Biomolecules. 2021;11(6):886. doi: 10.3390/biom11060886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  313. Nagalingam M., Kalpana V. N., Rajeswari V. D., Panneerselvam A.. Biosynthesis, Characterization, and Evaluation of Bioactivities of Leaf Extract-Mediated Biocompatible Gold Nanoparticles from Alternanthera Bettzickiana. Biotechnol. Rep. 2018;19:e00268. doi: 10.1016/j.btre.2018.e00268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Lima E., Guerra R., Lara V., Guzmán A.. Gold Nanoparticles as Efficient Antimicrobial Agents for Escherichia Coli and Salmonella Typhi. Chem. Cent. J. 2013;7(1):11. doi: 10.1186/1752-153X-7-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Li X., Robinson S. M., Gupta A., Saha K., Jiang Z., Moyano D. F., Sahar A., Riley M. A., Rotello V. M.. Functional Gold Nanoparticles as Potent Antimicrobial Agents against Multi-Drug-Resistant Bacteria. ACS Nano. 2014;8(10):10682–10686. doi: 10.1021/nn5042625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  316. Timoszyk A., Grochowalska R.. Mechanism and Antibacterial Activity of Gold Nanoparticles (AuNPs) Functionalized with Natural Compounds from Plants. Pharmaceutics. 2022;14:2599. doi: 10.3390/pharmaceutics14122599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. El-Khawaga A. M., Elsaidy A., Correa-Duarte M. A., Elbasuney S.. Unveiling the Photocatalytic and Antimicrobial Activities of Star–Shaped Gold Nanoparticles under Visible Spectrum. Sci. Rep. 2025;15(1):1201. doi: 10.1038/s41598-024-82332-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  318. Aati S., Aati H. Y., Hamed A. A., El-Shamy S., Aati S. H., Abdelmohsen U. R., Bringmann G., Taha M. N., Hassan H. M., Bahr H. S.. Gold Nanoparticles Synthesized from Soil-Derived Streptomyces Sp. ASM19: Characterization, Antimicrobial, Anticancer Potency Targeted G2/M Phase Cell-Cycle Arrest, and in Silico Studies. RSC Adv. 2025;15(5):3954–3968. doi: 10.1039/D4RA07608G. [DOI] [PMC free article] [PubMed] [Google Scholar]
  319. Alsareii, S. A. ; Alamri, A. M. ; AlWadai, H. H. A. ; Mahnashi, M. H. ; Shaikh, I. A. ; Shettar, A. K. ; Hoskeri, J. H. ; Mannasaheb, B. A. . Cassia Sericea Leaf Extract for Eco-Friendly Synthesis of Silver and Gold Nanoparticles with Boosted Wound Healing and Antimicrobial Potential Arabian J. Sci. Eng. 2025. 10.1007/s13369-025-09988-w. [DOI]
  320. Zhao Y., Zhan K., Geng P., Jiang S.. Polydopamine-Assisted Decoration of Silver Nanoparticles on Gold Nanorods for Photothermal and Chemical Antimicrobial Applications. New J. Chem. 2025;49(2):624–631. doi: 10.1039/D4NJ04434G. [DOI] [Google Scholar]
  321. Liu R., Wang P., Chen Y., Huang F., Shen Y., Zheng Y., Zheng L.. Fluorescent Sensor Array for Rapid Bacterial Identification Using Antimicrobial Peptide-Functionalized Gold Nanoclusters and Machine Learning. Talanta. 2025;291:127883. doi: 10.1016/j.talanta.2025.127883. [DOI] [PubMed] [Google Scholar]
  322. Gates K., Rai S., Kolawole O. P., Kundu S., Pramanik A., Singh S., Bandari P., Pandey V., Morehead D., Alamgir R., Edorodion Z., Dinadayalane T., Ray P. C.. Photothermal-Photocatalytic Ternary Heterostructure for Solar Light-Driven Highly Efficient Degradation of Antimicrobial Agents and Inactivation of Superbugs. ACS Appl. Bio Mater. 2025;8:1732–1744. doi: 10.1021/acsabm.4c01948. [DOI] [PubMed] [Google Scholar]
  323. Vostrejs P., Kovalcik A., Schaubeder J. B., Spirk S., Hricovini M., Nypelö T., Kalina M., Cernekova N., Brezina M., Kargl R.. Biobased Antimicrobial Coatings: Layer-by-Layer Assemblies Based on Natural Polyphenols. ACS Appl. Polym. Mater. 2025;7:3601–3610. doi: 10.1021/acsapm.4c03681. [DOI] [Google Scholar]
  324. Cao T., Zhou Y., Zhang J., Zhao B.. Clean Preparation of Ag@Au Nanoparticle-Modified Cotton Fabric with Durable Antimicrobial Properties by Adsorption. Cellulose. 2025;32:613–628. doi: 10.1007/s10570-024-06299-5. [DOI] [Google Scholar]
  325. Kaliappan K., Nagarajan P., Jayabalan J., Pushparaj H., Elumalai S., Paramanathan B., Manickam V., Jang H. T., Mani G.. Systematic Antimicrobial, Biofilm, Free Radical Inhibition and Tyrosinase Inhibition Assessments of Efficient Green Silver Nanoparticles from the Aqueous Root Extract of Cyphostemma Adenocaule (CA) RSC Pharm. 2025;2(1):147–162. doi: 10.1039/D4PM00173G. [DOI] [Google Scholar]
  326. Oh D., Khan F., Park S.-K., Jo D.-M., Kim N.-G., Jung W.-K., Kim Y.-M.. Antimicrobial, Antibiofilm, and Antivirulence Properties of Eisenia Bicyclis-Extracts and Eisenia Bicyclis-Gold Nanoparticles towards Microbial Pathogens. Microb. Pathog. 2024;188:106546. doi: 10.1016/j.micpath.2024.106546. [DOI] [PubMed] [Google Scholar]
  327. Suliasih B. A., Budi S., Katas H.. et al. Synthesis and Application of Gold Nanoparticles as Antioxidants. Pharmacia. 2024;71:1–19. doi: 10.3897/pharmacia.71.e112322. [DOI] [Google Scholar]
  328. Seku K., Reddy G. B., Osman A. I., Hussaini S. S., Kumar N. S., Al-Abri M., Pejjai B., Alreshaidan S. B., Al-Fatesh A. S., Kadimpati K. K.. Modified Frankincense Resin Stabilized Gold Nanoparticles for Enhanced Antioxidant and Synergetic Activity in In-Vitro Anticancer Studies. Int. J. Biol. Macromol. 2024;278:134935. doi: 10.1016/j.ijbiomac.2024.134935. [DOI] [PubMed] [Google Scholar]
  329. Experimental-Relations-of-Gold-(and-Other-Metals)-to-Light.
  330. Suliasih B. A., Sakinah A., Angelina M.. The Effect of Electrodeposition Voltage on the Antioxidant Activity of Gold Nanoparticles. Chem. Mater. 2024;3(1):27–33. doi: 10.56425/cma.v3i1.72. [DOI] [Google Scholar]
  331. Aamir M., Hassan S., Khan A. H., Ibrar M., Sarwar S., Mahmood K., Khan N., Aljumaiah D. A., Aldiaram A. H., Alameer A. K., Alsalman A. J., Farid A.. Spirulina-Mediated Biosynthesis of Gold Nanoparticles: An Interdisciplinary Study on Antimicrobial, Antioxidant, and Anticancer Properties. J. Solgel Sci. Technol. 2025;113:749. doi: 10.1007/s10971-024-06649-6. [DOI] [Google Scholar]
  332. Pu S., Li J., Sun L., Zhong L., Ma Q.. An in Vitro Comparison of the Antioxidant Activities of Chitosan and Green Synthesized Gold Nanoparticles. Carbohydr. Polym. 2019;211:161–172. doi: 10.1016/j.carbpol.2019.02.007. [DOI] [PubMed] [Google Scholar]
  333. Al-Radadi N. S.. Synthesis, Characterization, Antimalarial, Antimicrobial, and Antioxidant Activities of GNPs with Annona Muricata Leaf Extract. Arabian J. Chem. 2025;18(1):106081. doi: 10.1016/j.arabjc.2024.106081. [DOI] [Google Scholar]
  334. Shunmugam R., Balusamy S. R., Kumar V., Menon S., Lakshmi T., Perumalsamy H.. Biosynthesis of Gold Nanoparticles Using Marine Microbe (Vibrio Alginolyticus) and Its Anticancer and Antioxidant Analysis. J. King Saud Univ. Sci. 2021;33(1):101260. doi: 10.1016/j.jksus.2020.101260. [DOI] [Google Scholar]
  335. Ameen F., Al-Maary K. S., Almansob A., AlNadhari S.. Antioxidant, Antibacterial and Anticancer Efficacy of Alternaria Chlamydospora-Mediated Gold Nanoparticles. Appl. Nanosci. 2023;13(3):2233–2240. doi: 10.1007/s13204-021-02047-4. [DOI] [Google Scholar]
  336. Mikhailova E. O.. Gold Nanoparticles: Biosynthesis and Potential of Biomedical Application. J. Funct. Biomater. 2021;12:70. doi: 10.3390/jfb12040070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  337. Hussien A. G., Aboulthana W. M., Youssef A. M.. In Vitro Evaluation of Saussurea Costus Gold Nano-Extract as Antioxidant, Anti-Diabetic, Anti-Alzheimer, and Anti-Inflammatory Agent. Egypt J. Chem. 2024;67(5):257–283. doi: 10.21608/EJCHEM.2023.244987.8786. [DOI] [Google Scholar]
  338. Ningaraju S., Munawer U., Raghavendra V. B., Balaji K. S., Melappa G., Brindhadevi K., Pugazhendhi A.. Chaetomium Globosum Extract Mediated Gold Nanoparticle Synthesis and Potent Anti-Inflammatory Activity. Anal. Biochem. 2021;612:113970. doi: 10.1016/j.ab.2020.113970. [DOI] [PubMed] [Google Scholar]
  339. Sivakavinesan M., Vanaja M., Annadurai G., Murugan A. M., Salmen S. H., Aljawdah H. M.. Antioxidant and Anti-Inflammatory Activity of Gold Nanoparticles Synthesized Employing Fruit Peel Extract of Citrus Sinensis L.Osbeck. Mater. Res. Express. 2024;11(10):105001. doi: 10.1088/2053-1591/ad7dd4. [DOI] [Google Scholar]
  340. Gao L., Mei S., Ma H., Chen X.. Ultrasound-Assisted Green Synthesis of Gold Nanoparticles Using Citrus Peel Extract and Their Enhanced Anti-Inflammatory Activity. Ultrason. Sonochem. 2022;83:105940. doi: 10.1016/j.ultsonch.2022.105940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  341. Patra J. K., Shin H. S., Yang I. J., Nguyen L. T. H., Das G.. Sustainable Utilization of Food Biowaste (Papaya Peel) Extract for Gold Nanoparticle Biosynthesis and Investigation of Its Multi-Functional Potentials. Antioxidants. 2024;13(5):581. doi: 10.3390/antiox13050581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Eltahir A. O. E., Lategan K. L., David O. M., Pool E. J., Luckay R. C., Hussein A. A.. Green Synthesis of Gold Nanoparticles Using Liquiritin and Other Phenolics from Glycyrrhiza Glabra and Their Anti-Inflammatory Activity. J. Funct. Biomater. 2024;15(4):95. doi: 10.3390/jfb15040095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Nasab D. M., Taheri A., Athari S. S.. Evaluation Anti-Inflammatory Effect of Conjugated Gold Nanoparticles with Cortistatin Peptide as Drug Delivery to Asthmatic Lung Tissue. Int. J. Pept. Res. Ther. 2023;29(1):16. doi: 10.1007/s10989-022-10487-x. [DOI] [Google Scholar]
  344. M U., Prasad D. R., N S., Raghavendra V. B., H P S.. Studies of in Vitro Antioxidant and Anti-Inflammatory Activities of Gold Nanoparticles Biosynthesised from a Medicinal Plant, Commiphora Wightii. Mater. Technol. 2022;37(9):915–925. doi: 10.1080/10667857.2021.1905206. [DOI] [Google Scholar]
  345. Hongsa N., Thinbanmai T., Luesakul U., Sansanaphongpricha K., Muangsin N.. A Novel Modified Chitosan/Collagen Coated-Gold Nanoparticles for 5-Fluorouracil Delivery: Synthesis, Characterization, in Vitro Drug Release Studies, Anti-Inflammatory Activity and in Vitro Cytotoxicity Assay. Carbohydr. Polym. 2022;277:118858. doi: 10.1016/j.carbpol.2021.118858. [DOI] [PubMed] [Google Scholar]
  346. Chen Y., Feng X.. Gold Nanoparticles for Skin Drug Delivery. Int. J. Pharm. 2022;625:122122. doi: 10.1016/j.ijpharm.2022.122122. [DOI] [PubMed] [Google Scholar]
  347. Qiu L., Wang C., Lan M., Guo Q., Du X., Zhou S., Cui P., Hong T., Jiang P., Wang J., Xia J.. Antibacterial Photodynamic Gold Nanoparticles for Skin Infection. ACS Appl. Bio Mater. 2021;4(4):3124–3132. doi: 10.1021/acsabm.0c01505. [DOI] [PubMed] [Google Scholar]
  348. Cuyler M., Twilley D., Thipe V. C., Mandiwana V., Kalombo M. L., Ray S. S., Rikhotso-Mbungela R. S., van Vuuren A. J., Coetsee W., Katti K. V., Lall N.. Antihistamine and Wound Healing Potential of Gold Nanoparticles Synthesized Using Bulbine Frutescens (L.) Willd. Nanotechnol. Sci. Appl. 2024;17:59–75. doi: 10.2147/NSA.S445116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Dastgheib Z. S., Abolmaali S. S., Farahavar G., Salmanpour M., Tamaddon A. M.. Gold Nanostructures in Melanoma: Advances in Treatment, Diagnosis, and Theranostic Applications. Heliyon. 2024;10:e35655. doi: 10.1016/j.heliyon.2024.e35655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Leu J. G., Chen S. A., Chen H. M., Wu W. M., Hung C. F., Yao Y. Der., Tu C. S., Liang Y. J.. The Effects of Gold Nanoparticles in Wound Healing with Antioxidant Epigallocatechin Gallate and α-Lipoic Acid. Nanomedicine. 2012;8(5):767–775. doi: 10.1016/j.nano.2011.08.013. [DOI] [PubMed] [Google Scholar]
  351. de Oliveira D., Luiz G. P., Scussel R., Fagundes M. I., Galvani N. C., Abel J. da S., Zaccaron R. P., de Bem Silveira G., de Andrade T. A. M., Silveira P. C. L., Machado-de-Ávila R. A.. The Combined Treatment of Gold Nanoparticles Associated with Photobiomodulation Accelerate the Healing of Dermonecrotic Lesion. J. Drug Target. 2024;32(2):172–185. doi: 10.1080/1061186X.2023.2298848. [DOI] [PubMed] [Google Scholar]
  352. Poomrattanangoon S., Pissuwan D.. Gold Nanoparticles Coated with Collagen-I and Their Wound Healing Activity in Human Skin Fibroblast Cells. Heliyon. 2024;10(13):e33302. doi: 10.1016/j.heliyon.2024.e33302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Dong Y., Wang Z., Wang J., Sun X., Yang X., Liu G.. Mussel-Inspired Electroactive, Antibacterial and Antioxidative Composite Membranes with Incorporation of Gold Nanoparticles and Antibacterial Peptides for Enhancing Skin Wound Healing. J. Biol. Eng. 2024;18(1):3. doi: 10.1186/s13036-023-00402-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  354. Wu X., Lu Y., Gao Y., Kang J., Dong A.. Gold Nanoparticles-Based Photothermal Hydrogel Assisted by N-Halamine for Bacterial-Infected Skin Wound Healing. Nanoscale. 2024;16:18348–18355. doi: 10.1039/D4NR02694B. [DOI] [PubMed] [Google Scholar]
  355. Abbas H. A., Taha A. A., Sulaiman G. M., Al Ali A., Al Shmrany H., Stamatis H., Mohammed H. A., Khan R. A.. Efficacy of Alginate-Coated Gold Nanoparticles against Antibiotics-Resistant Staphylococcus and Streptococcus Pathogens of Acne Origins. Open Life Sci. 2025;20(1):20221045. doi: 10.1515/biol-2022-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  356. Ko W. C., Wang S. J., Hsiao C. Y., Hung C. T., Hsu Y. J., Chang D. C., Hung C. F.. Pharmacological Role of Functionalized Gold Nanoparticles in Disease Applications. Molecules. 2022;27:1551. doi: 10.3390/molecules27051551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Apaolaza P. S., Busch M., Asin-Prieto E., Peynshaert K., Rathod R., Remaut K., Dünker N., Göpferich A.. Hyaluronic Acid Coating of Gold Nanoparticles for Intraocular Drug Delivery: Evaluation of the Surface Properties and Effect on Their Distribution. Exp. Eye Res. 2020;198:108151. doi: 10.1016/j.exer.2020.108151. [DOI] [PubMed] [Google Scholar]
  358. Sonntag T., Froemel F., Stamer W. D., Ohlmann A., Fuchshofer R., Breunig M.. Distribution of Gold Nanoparticles in the Anterior Chamber of the Eye after Intracameral Injection for Glaucoma Therapy. Pharmaceutics. 2021;13(6):901. doi: 10.3390/pharmaceutics13060901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Milan J., Niemczyk K., Kus-Liśkiewicz M.. Treasure on the EarthGold Nanoparticles and Their Biomedical Applications. Materials. 2022;15:3355. doi: 10.3390/ma15093355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Kavalaraki A., Spyratou E., Kouri M. A., Efstathopoulos E. P.. Gold Nanoparticles as Contrast Agents in Ophthalmic Imaging. Optics. 2023;4:74–99. doi: 10.3390/opt4010007. [DOI] [Google Scholar]
  361. Kuzuya T., Nakagawa S., Satoh J., Kanazawa Y., Iwamoto Y., Kobayashi M., Nanjo K., Sasaki A., Seino Y., Ito C., Shima K., Nonaka K., Kadowaki T.. Report of the Committee on the Classification and Diagnostic Criteria of Diabetes Mellitus. Diabetes Res. Clin. Pract. 2002;55(1):65–85. doi: 10.1016/S0168-8227(01)00365-5. [DOI] [PubMed] [Google Scholar]
  362. Subramani K., Pathak S., Hosseinkhani H.. Recent trends in diabetes treatment using nanotechnologY. Digest J. Nanomater. Biostruct. 2012;7:85–95. [Google Scholar]
  363. Alomari G., Hamdan S., Al-Trad B.. Gold Nanoparticles as a Promising Treatment for Diabetes and Its Complications: Current and Future Potentials. Braz. J. Pharm. Sci. 2021;57:e19040. doi: 10.1590/s2175-97902020000419040. [DOI] [Google Scholar]
  364. Daisy P., Saipriya K.. Biochemical Analysis of Cassia Fistula Aqueous Extract and Phytochemically Synthesized Gold Nanoparticles as Hypoglycemic Treatment for Diabetes Mellitus. Int. J. Nanomed. 2012;7:1189–1202. doi: 10.2147/IJN.S26650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Al-Shwaheen A., Aljabali A. A. A., Alomari G., Al Zoubi M., Alshaer W., Al-Trad B., Tambuwala M. M.. Molecular and Cellular Effects of Gold Nanoparticles Treatment in Experimental Diabetic Myopathy. Heliyon. 2022;8(9):e10358. doi: 10.1016/j.heliyon.2022.e10358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Opris R., Tatomir C., Olteanu D., Moldovan R., Moldovan B., David L., Nagy A., Decea N., Kiss M. L., Filip G. A.. The Effect of Sambucus Nigra L. Extract and Phytosinthesized Gold Nanoparticles on Diabetic Rats. Colloids Surf., B. 2017;150:192–200. doi: 10.1016/j.colsurfb.2016.11.033. [DOI] [PubMed] [Google Scholar]
  367. Younis N. K., Ghoubaira J. A., Bassil E. P., Tantawi H. N., Eid A. H.. Metal-Based Nanoparticles: Promising Tools for the Management of Cardiovascular Diseases. Nanomedicine. 2021;36:102433. doi: 10.1016/j.nano.2021.102433. [DOI] [PubMed] [Google Scholar]
  368. Wu Y., Vazquez-Prada K. X., Liu Y., Whittaker A. K., Zhang R., Ta H. T.. Recent Advances in the Development of Theranostic Nanoparticles for Cardiovascular Diseases. Nanotheranostics. 2021;5:99–514. doi: 10.7150/ntno.62730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  369. Soumya R. S., Raghu K. G.. Recent Advances on Nanoparticle-Based Therapies for Cardiovascular Diseases. J. Cardiol. 2023;81:10–18. doi: 10.1016/j.jjcc.2022.02.009. [DOI] [PubMed] [Google Scholar]
  370. Adv Healthcare Materials2023RoshanbinfarCollagen Hydrogel Containing Polyethylenimine-Gold Nanoparticles for Drug.Pdf.
  371. Luo R., Li J., Huang G., Li G., Guo S., Yuan Y.. Electrochemical Biosensor for the Detection of Low Density Lipoprotein Based on Gold Nanoparticles Mediated Bi-Enzymes Catalytic Silver Deposition Reaction. Microchem. J. 2024;199:109927. doi: 10.1016/j.microc.2024.109927. [DOI] [Google Scholar]
  372. van der Ven C. F. T., Tibbitt M. W., Conde J., van Mil A., Hjortnaes J., Doevendans P. A., Sluijter J. P. G., Aikawa E., Langer R. S.. Controlled Delivery of Gold Nanoparticle-Coupled MiRNA Therapeutics via an Injectable Self-Healing Hydrogel. Nanoscale. 2021;13(48):20451–20461. doi: 10.1039/D1NR04973A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Tu L., Zou Z., Yang Y., Wang S., Xing B., Feng J., Jin Y., Cheng M.. Targeted Drug Delivery Systems for Atherosclerosis. J. Nanobiotechnol. 2025;23(1):306. doi: 10.1186/s12951-025-03384-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  374. Khan S., Hasan A., Attar F., Sharifi M., Siddique R., Mraiche F., Falahati M.. Gold Nanoparticle-Based Platforms for Diagnosis and Treatment of Myocardial Infarction. ACS Biomater. Sci. Eng. 2020;6(12):6460–6477. doi: 10.1021/acsbiomaterials.0c00955. [DOI] [PubMed] [Google Scholar]
  375. Silva I. V. G., de Figueiredo R. C., Alves Rios D. R.. Effect of Different Classes of Antihypertensive Drugs on Endothelial Function and Inflammation. Int. J. Mol. Sci. 2019;20:3458. doi: 10.3390/ijms20143458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Lv J., Zhang L., Du W., Ling G., Zhang P.. Functional Gold Nanoparticles for Diagnosis, Treatment and Prevention of Thrombus. J. Controlled Release. 2022;345:572–585. doi: 10.1016/j.jconrel.2022.03.044. [DOI] [PubMed] [Google Scholar]
  377. Wang Z., Yang N., Hou Y., Li Y., Yin C., Yang E., Cao H., Hu G., Xue J., Yang J., Liao Z., Wang W., Sun D., Fan C., Zheng L.. L-Arginine-Loaded Gold Nanocages Ameliorate Myocardial Ischemia/Reperfusion Injury by Promoting Nitric Oxide Production and Maintaining Mitochondrial Function. Adv. Sci. 2023;10(26):2302123. doi: 10.1002/advs.202302123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Nikolaou K., Alkadhi H., Bamberg F., Leschka S., Wintersperger B. J.. MRI and CT in the Diagnosis of Coronary Artery Disease: Indications and Applications. Insights into Imaging. 2011;2:9–24. doi: 10.1007/s13244-010-0049-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  379. Dela Rosa J. G. L., Bantayan N. R. B., Tantengco O. A. G.. Recent Developments in Nanoparticle-Based Treatment for Atrial Fibrillation and Other Human Cardiac Arrhythmias. J. Cardiovasc. Aging. 2025;5(2):9. doi: 10.20517/jca.2024.25. [DOI] [Google Scholar]
  380. Chiang M. C., Yang Y. P., Nicol C. J. B., Wang C. J.. Gold Nanoparticles in Neurological Diseases: A Review of Neuroprotection. Int. J. Mol. Sci. 2024;25:2360. doi: 10.3390/ijms25042360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Lv J., Zhang L., Du W., Ling G., Zhang P.. Functional Gold Nanoparticles for Diagnosis, Treatment and Prevention of Thrombus. J. Controlled Release. 2022;345:572–585. doi: 10.1016/j.jconrel.2022.03.044. [DOI] [PubMed] [Google Scholar]
  382. Sabir F., Barani M., Mukhtar M., Rahdar A., Cucchiarini M., Zafar M. N., Behl T., Bungau S.. Nanodiagnosis and Nanotreatment of Cardiovascular Diseases: An Overview. Chemosensors. 2021;9:67. doi: 10.3390/chemosensors9040067. [DOI] [Google Scholar]
  383. Agnihotri V., Agrawal Y., Goyal S., Sharma C., Ojha S.. An Update on Advancements and Challenges in Inhalational Drug Delivery for Pulmonary Arterial Hypertension. Molecules. 2022;27(11):3490. doi: 10.3390/molecules27113490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  384. Milan J., Niemczyk K., Kus-Liśkiewicz M.. Treasure on the Earth-Gold Nanoparticles and Their Biomedical Applications. Materials. 2022;15:3355. doi: 10.3390/ma15093355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  385. Sun L., Liu H., Ye Y., Lei Y., Islam R., Tan S., Tong R., Miao Y.-B., Cai L.. Smart Nanoparticles for Cancer Therapy. Signal Transduction Targeted Ther. 2023;8(1):418. doi: 10.1038/s41392-023-01642-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Tomar A., Garg G.. Short Review on Application of Gold Nanoparticles. Global J. Pharmacol. 2013;7(1):34–38. doi: 10.5829/idosi.gjp.2013.7.1.66173. [DOI] [Google Scholar]
  387. Raoof M., Curley S. A.. Non-Invasive Radiofrequency-Induced Targeted Hyperthermia for the Treatment of Hepatocellular Carcinoma. Int. J. Hepatol. 2011;2011:676957. doi: 10.4061/2011/676957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Dai X., Li X., Du Y., Han M., Wang Z., Wang Y., Yan F., Liu Y.. Gold Nanorod–Mesoporous Silica Core Shell Nanocomposites for NIR-II Photothermal Ablation and Dual PD-L1/VEGF Blockade Therapy in Hepatocellular Carcinoma. Chem. Eng. J. 2023;459:141426. doi: 10.1016/j.cej.2023.141426. [DOI] [Google Scholar]
  389. Ahmadi S. M., Seyedabadi M., Ebrahimnejad P., Abasi M., Nokhodchi A.. Efficient Delivery of Gold Nanoparticles and MiRNA-33a Via Cationic PEGylated Niosomal Formulation to MCF-7 Breast Cancer Cells. AAPS PharmSciTech. 2024;25(7):213. doi: 10.1208/s12249-024-02906-7. [DOI] [PubMed] [Google Scholar]
  390. Tu L., Luo Z., Wu Y. L., Huo S., Liang X. J.. Gold-Based Nanomaterials for the Treatment of Brain Cancer. Cancer Biol. Med. 2021;18:372–387. doi: 10.20892/j.issn.2095-3941.2020.0524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  391. Lee D. G., Lee M., Go E. B., Chung N.. Resveratrol-Loaded Gold Nanoparticles Enhance Caspase-Mediated Apoptosis in PANC-1 Pancreatic Cells via Mitochondrial Intrinsic Apoptotic Pathway. Cancer Nanotechnol. 2022;13(1):34. doi: 10.1186/s12645-022-00143-w. [DOI] [Google Scholar]
  392. Nguyen P. V., Allard-Vannier E., Chourpa I., Hervé-Aubert K.. Nanomedicines Functionalized with Anti-EGFR Ligands for Active Targeting in Cancer Therapy: Biological Strategy, Design and Quality Control. Int. J. Pharm. 2021;605:120795. doi: 10.1016/j.ijpharm.2021.120795. [DOI] [PubMed] [Google Scholar]
  393. Yang W., Liang H., Ma S., Wang D., Huang J.. Gold Nanoparticle Based Photothermal Therapy: Development and Application for Effective Cancer Treatment. Sustainable Mater. Technol. 2019;22:e00109. doi: 10.1016/j.susmat.2019.e00109. [DOI] [Google Scholar]
  394. Khoshfetrat S. M., Mehrgardi M. A.. Amplified Detection of Leukemia Cancer Cells Using an Aptamer-Conjugated Gold-Coated Magnetic Nanoparticles on a Nitrogen-Doped Graphene Modified Electrode. Bioelectrochemistry. 2017;114:24–32. doi: 10.1016/j.bioelechem.2016.12.001. [DOI] [PubMed] [Google Scholar]
  395. Mahmoud N. N., Salman T. M., Al-Dabash S., Abdullah M., Abu-Dahab R.. The Impact of Gold Nanoparticles Conjugated with Albumin on Prostate and Breast Cancer Cell Lines: Insights into Cytotoxicity, Cellular Uptake, Migration, and Adhesion Potential. J. Nanopart. Res. 2024;26(5):101. doi: 10.1007/s11051-024-05990-9. [DOI] [Google Scholar]
  396. Sonkar R., Sonali, Jha A., Viswanadh M. K., Burande A. S., Narendra, Pawde D. M., Patel K. K., Singh M., Koch B., Muthu M. S.. Gold Liposomes for Brain-Targeted Drug Delivery: Formulation and Brain Distribution Kinetics. Mater. Sci. Eng.: C. 2021;120:111652. doi: 10.1016/j.msec.2020.111652. [DOI] [PubMed] [Google Scholar]
  397. Alric C., Hervé-Aubert K., Aubrey N., Melouk S., Lajoie L., Même W., Même S., Courbebaisse Y., Ignatova A. A., Feofanov A. V., Chourpa I., Allard-Vannier E.. Targeting HER2-Breast Tumors with ScFv-Decorated Bimodal Nanoprobes. J. Nanobiotechnol. 2018;16(1):18. doi: 10.1186/s12951-018-0341-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  398. Portela, M. G. ; Pedrouzo, L. ; Abuin, C. ; López, R. ; Alonso, L. ; Cernadas, A. ; Ferreirós, A. ; Dávila Ibáñez, A. B. . HER2-Gold Nanoparticles Platform to Detect and Isolate Positive Exosomes and Select HER2-Positive Breast Cancer Patients More Accurately. https://ssrn.com/abstract=5270229.
  399. Alafaleq N. O., Alomari A., Khan M. S., Shaik G. M., Hussain A., Ahmed F., Hassan I., M Alhazza I., Alokail M. S., Alenad A. M. H., Jabir N. R., Tabrez S.. Anticancer Potential of Gold Nanoparticles (AuNPs) Using a Battery of in Vitro Tests. Nanotechnol. Rev. 2022;11(1):3292–3304. doi: 10.1515/ntrev-2022-0502. [DOI] [Google Scholar]
  400. Balasubramani G., Ramkumar R., Krishnaveni N., Pazhanimuthu A., Natarajan T., Sowmiya R., Perumal P.. Structural Characterization, Antioxidant and Anticancer Properties of Gold Nanoparticles Synthesized from Leaf Extract (Decoction) of Antigonon Leptopus Hook. & Arn. J. Trace Elem. Med. Biol. 2015;30:83–89. doi: 10.1016/j.jtemb.2014.11.001. [DOI] [PubMed] [Google Scholar]
  401. Botteon C. E. A., Silva L. B., Ccana-Ccapatinta G. V., Silva T. S., Ambrosio S. R., Veneziani R. C. S., Bastos J. K., Marcato P. D.. Biosynthesis and Characterization of Gold Nanoparticles Using Brazilian Red Propolis and Evaluation of Its Antimicrobial and Anticancer Activities. Sci. Rep. 2021;11(1):1974. doi: 10.1038/s41598-021-81281-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Zhang P., Wang P., Yan L., Liu L.. Synthesis of Gold Nanoparticles with Solanum Xanthocarpum Extract and Their in Vitro Anticancer Potential on Nasopharyngeal Carcinoma Cells. Int. J. Nanomed. 2018;13:7047–7059. doi: 10.2147/IJN.S180138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Steckiewicz K. P., Barcinska E., Malankowska A., Zauszkiewicz–Pawlak A., Nowaczyk G., Zaleska-Medynska A., Inkielewicz-Stepniak I.. Impact of Gold Nanoparticles Shape on Their Cytotoxicity against Human Osteoblast and Osteosarcoma in in Vitro Model. Evaluation of the Safety of Use and Anti-Cancer Potential. J. Mater. Sci. Mater. Med. 2019;30(2):22. doi: 10.1007/s10856-019-6221-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Vemuri S. K., Banala R. R., Mukherjee S., Uppula P., GPV S., A V G. R., T M.. Novel Biosynthesized Gold Nanoparticles as Anti-Cancer Agents against Breast Cancer: Synthesis, Biological Evaluation, Molecular Modelling Studies. Mater. Sci. Eng.: C. 2019;99:417–429. doi: 10.1016/j.msec.2019.01.123. [DOI] [PubMed] [Google Scholar]
  405. Donga S., Bhadu G. R., Chanda S.. Antimicrobial, Antioxidant and Anticancer Activities of Gold Nanoparticles Green Synthesized Using Mangifera Indica Seed Aqueous Extract. Artif. Cells Nanomed. Biotechnol. 2020;48(1):1315–1325. doi: 10.1080/21691401.2020.1843470. [DOI] [PubMed] [Google Scholar]
  406. Ajdari Z., Rahman H., Shameli K., Abdullah R., Abd Ghani M., Yeap S., Abbasiliasi S., Ajdari D., Ariff A.. Novel Gold Nanoparticles Reduced by Sargassum Glaucescens: Preparation, Characterization and Anticancer Activity. Molecules. 2016;21(3):123. doi: 10.3390/molecules21030123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Dheyab M. A., Aziz A. A., Al-Mafarjy S. S., Suardi N., Razak N. N. A. N. A., Ramizy A., Jameel M. S.. Exploring the Anticancer Potential of Biogenic Inorganic Gold Nanoparticles Synthesized via Mushroom-Assisted Green Route. Inorg. Chem. Commun. 2023;157:111363. doi: 10.1016/j.inoche.2023.111363. [DOI] [Google Scholar]
  408. Mamman A., Jain P., Srivastava V.. Exploring the Catalytic, Bactericidal, and Anticancer Potential of Ag0, Au0, and Ag0@Au0 Nanoparticles: A Comparative Study. Colloids Surf., A. 2025;707:135855. doi: 10.1016/j.colsurfa.2024.135855. [DOI] [Google Scholar]
  409. Susanna D., Balakrishnan R. M., Ponnan Ettiyappan J.. Ultrasonication-Assisted Green Synthesis and Characterization of Gold Nanoparticles from Nothapodytes Foetida: An Assessment of Their Antioxidant, Antibacterial, Anticancer and Wound Healing Potential. J. Drug Delivery Sci. Technol. 2023;87:104740. doi: 10.1016/j.jddst.2023.104740. [DOI] [Google Scholar]
  410. Umapathi A., PN N., Madhyastha H., Singh M., Madhyastha R., Maruyama M., Daima H. K.. Curcumin and Isonicotinic Acid Hydrazide Functionalized Gold Nanoparticles for Selective Anticancer Action. Colloids Surf., A. 2020;607:125484. doi: 10.1016/j.colsurfa.2020.125484. [DOI] [Google Scholar]
  411. Wang L., Xu J., Yan Y., Liu H., Li F.. Synthesis of Gold Nanoparticles from Leaf Panax Notoginseng and Its Anticancer Activity in Pancreatic Cancer PANC-1 Cell Lines. Artif. Cells Nanomed. Biotechnol. 2019;47(1):1216–1223. doi: 10.1080/21691401.2019.1593852. [DOI] [PubMed] [Google Scholar]
  412. Esther Lydia D., Khusro A., Immanuel P., Esmail G. A., Al-Dhabi N. A., Arasu M. V.. Photo-Activated Synthesis and Characterization of Gold Nanoparticles from Punica Granatum L. Seed Oil: An Assessment on Antioxidant and Anticancer Properties for Functional Yoghurt Nutraceuticals. J. Photochem. Photobiol. B. 2020;206:111868. doi: 10.1016/j.jphotobiol.2020.111868. [DOI] [PubMed] [Google Scholar]
  413. Asl S. S., Tafvizi F., Noorbazargan H.. Biogenic Synthesis of Gold Nanoparticles Using Satureja Rechingeri Jamzad: A Potential Anticancer Agent against Cisplatin-Resistant A2780CP Ovarian Cancer Cells. Environ. Sci. Pollut. Res. 2023;30(8):20168–20184. doi: 10.1007/s11356-022-23507-6. [DOI] [PubMed] [Google Scholar]
  414. Kiran M. S., Kumar C. R., Shwetha U. R., Onkarappa H. S., Betageri V. S., Latha M. S.. Green Synthesis and Characterization of Gold Nanoparticles from Moringa Oleifera Leaves and Assessment of Antioxidant, Antidiabetic and Anticancer Properties. Chem. Data Collect. 2021;33:100714. doi: 10.1016/j.cdc.2021.100714. [DOI] [Google Scholar]
  415. Padalia H., Chanda S.. Antioxidant and Anticancer Activities of Gold Nanoparticles Synthesized Using Aqueous Leaf Extract of Ziziphus Nummularia. Bionanoscience. 2021;11(2):281–294. doi: 10.1007/s12668-021-00849-y. [DOI] [Google Scholar]
  416. Akrami M., Samimi S., Alipour M., Bardania H., Ramezanpour S., Najafi N., Hosseinkhani S., Kamankesh M., Haririan I., Hassanshahi F.. Potential Anticancer Activity of a New Pro-Apoptotic Peptide–Thioctic Acid Gold Nanoparticle Platform. Nanotechnology. 2021;32(14):145101. doi: 10.1088/1361-6528/abd3cb. [DOI] [PubMed] [Google Scholar]
  417. Daei S., Ziamajidi N., Abbasalipourkabir R., Khanaki K., Bahreini F.. Anticancer Effects of Gold Nanoparticles by Inducing Apoptosis in Bladder Cancer 5637 Cells. Biol. Trace Elem. Res. 2022;200(6):2673–2683. doi: 10.1007/s12011-021-02895-9. [DOI] [PubMed] [Google Scholar]
  418. Qian L., Su W., Wang Y., Dang M., Zhang W., Wang C.. Synthesis and Characterization of Gold Nanoparticles from Aqueous Leaf Extract of Alternanthera Sessilis and Its Anticancer Activity on Cervical Cancer Cells (HeLa) Artif. Cells Nanomed. Biotechnol. 2019;47(1):1173–1180. doi: 10.1080/21691401.2018.1549064. [DOI] [PubMed] [Google Scholar]
  419. Al-Mafarjy S. S., Suardi N., Ahmed N. M., Kernain D., Hisham Alkatib H., Dheyab M. A.. Green Synthesis of Gold Nanoparticles from Coleus Scutellarioides (L.) Benth Leaves and Assessment of Anticancer and Antioxidant Properties. Inorg. Chem. Commun. 2024;161:112052. doi: 10.1016/j.inoche.2024.112052. [DOI] [Google Scholar]
  420. Fratoddi I., Venditti I., Battocchio C., Carlini L., Amatori S., Porchia M., Tisato F., Bondino F., Magnano E., Pellei M., Santini C.. Highly Hydrophilic Gold Nanoparticles as Carrier for Anticancer Copper­(I) Complexes: Loading and Release Studies for Biomedical Applications. Nanomaterials. 2019;9(5):772. doi: 10.3390/nano9050772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  421. Maity R., Chatterjee M., Banerjee A., Das A., Mishra R., Mazumder S., Chanda N.. Gold Nanoparticle-Assisted Enhancement in the Anti-Cancer Properties of Theaflavin against Human Ovarian Cancer Cells. Mater. Sci. Eng.: C. 2019;104:109909. doi: 10.1016/j.msec.2019.109909. [DOI] [PubMed] [Google Scholar]
  422. Samal S., Meher R. K., Das P. K., Swain S. K., Dubey D., Khan M. S., Jali B. R.. Exploring the Anticancer and Antioxidant Potential of Gold Nanoparticles Synthesized from Pterocarpus Marsupium Bark Extract against Oral Squamous Cell Carcinoma. Artif. Cells Nanomed. Biotechnol. 2024;52(1):512–528. doi: 10.1080/21691401.2024.2416951. [DOI] [PubMed] [Google Scholar]
  423. Dorosti N., Jamshidi F.. Plant-Mediated Gold Nanoparticles by Dracocephalum Kotschyi as Anticholinesterase Agent: Synthesis, Characterization, and Evaluation of Anticancer and Antibacterial Activity. J. Appl. Biomed. 2016;14(3):235–245. doi: 10.1016/j.jab.2016.03.001. [DOI] [Google Scholar]
  424. Perveen K., Husain F. M., Qais F. A., Khan A., Razak S., Afsar T., Alam P., Almajwal A. M., Abulmeaty M. M. A.. Microwave-Assisted Rapid Green Synthesis of Gold Nanoparticles Using Seed Extract of Trachyspermum Ammi: ROS Mediated Biofilm Inhibition and Anticancer Activity. Biomolecules. 2021;11(2):197. doi: 10.3390/biom11020197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. Zheng Y., Zhang J., Zhang R., Luo Z., Wang C., Shi S.. Gold Nano Particles Synthesized from Magnolia Officinalis and Anticancer Activity in A549 Lung Cancer Cells. Artif. Cells Nanomed. Biotechnol. 2019;47(1):3101–3109. doi: 10.1080/21691401.2019.1645152. [DOI] [PubMed] [Google Scholar]
  426. Baran M. F., Keskin C., Baran A., Eftekhari A., Omarova S., Khalilov R., Adican M. T., Rosić G., Selakovic D., Yıldıztekin M., Kurt K., Aytuğ Ava C., Atalar M. N.. The Investigation of the Chemical Composition and Applicability of Gold Nanoparticles Synthesized with Amygdalus Communis (Almond) Leaf Aqueous Extract as Antimicrobial and Anticancer Agents. Molecules. 2023;28(6):2428. doi: 10.3390/molecules28062428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  427. Adewale O. B., Anadozie S. O., Potts-Johnson S. S., Onwuelu J. O., Obafemi T. O., Osukoya O. A., Fadaka A. O., Davids H., Roux S.. Investigation of Bioactive Compounds in Crassocephalum Rubens Leaf and in Vitro Anticancer Activity of Its Biosynthesized Gold Nanoparticles. Biotechnol. Rep. 2020;28:e00560. doi: 10.1016/j.btre.2020.e00560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  428. Doghish A. S., Hashem A. H., Shehabeldine A. M., Sallam A.-A. M., El-Sayyad G. S., Salem S. S.. Nanocomposite Based on Gold Nanoparticles and Carboxymethyl Cellulose: Synthesis, Characterization, Antimicrobial, and Anticancer Activities. J. Drug Delivery Sci. Technol. 2022;77:103874. doi: 10.1016/j.jddst.2022.103874. [DOI] [Google Scholar]
  429. Singh A. K., Tiwari R., Singh V. K., Singh P., Khadim S. R., Singh U., Laxmi, Srivastava V., Hasan S. H., Asthana R. K.. Green Synthesis of Gold Nanoparticles from Dunaliella Salina, Its Characterization and in Vitro Anticancer Activity on Breast Cancer Cell Line. J. Drug Delivery Sci. Technol. 2019;51:164–176. doi: 10.1016/j.jddst.2019.02.023. [DOI] [Google Scholar]
  430. Shakoor A., Ferdous U. T., Khan S. A., Gulzar M. M.. Green Synthesis of Gold Nanoparticles Using Clerodendrum Trichotomum Thunberg for Antibacterial and Anticancer Applications. Int. J. Nanomedicine. 2025;20:2645–2658. doi: 10.2147/IJN.S503254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  431. Algotiml R., Gaballa A., Seoudi R., Abulreesh H. H., Ahmad I., Elbanna K.. Anticancer and Antimicrobial Activity of Red Sea Seaweeds Extracts-Mediated Gold Nanoparticles. J. Pure Appl. Microbiol. 2022;16(1):207–225. doi: 10.22207/JPAM.16.1.11. [DOI] [Google Scholar]
  432. Assad N., Laila M. B., Hassan M. N. U., Rehman M. F. ur., Ali L., Mustaqeem M., Ullah B., Khan M. N., Iqbal M., Ercişli S., Alarfaj A. A., Ansari M. J., Malik T.. Eco-Friendly Synthesis of Gold Nanoparticles Using Equisetum Diffusum D. Don. with Broad-Spectrum Antibacterial, Anticancer, Antidiabetic, and Antioxidant Potentials. Sci. Rep. 2025;15(1):19246. doi: 10.1038/s41598-025-02450-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Balashanmugam P., Sucharithra G., Mary S. A., Selvi T. SA.. Efficacy of Biopolymeric PVA-AuNPs and PCL-Curcumin Loaded Electrospun Nanofibers and Their Anticancer Activity against A431 Skin Cancer Cell Line. Mater. Today Commun. 2020;25:101276. doi: 10.1016/j.mtcomm.2020.101276. [DOI] [Google Scholar]
  434. Lee C.-S., Kim T. W., Oh D. E., Bae S. O., Ryu J., Kong H., Jeon H., Seo H. K., Jeon S., Kim T. H.. In Vivo and In Vitro Anticancer Activity of Doxorubicin-Loaded DNA-AuNP Nanocarrier for the Ovarian Cancer Treatment. Cancers. 2020;12(3):634. doi: 10.3390/cancers12030634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  435. Kumar P. S., Jeyalatha M. V., Malathi J., Ignacimuthu S.. Anticancer Effects of One-Pot Synthesized Biogenic Gold Nanoparticles (Mc-AuNps) against Laryngeal Carcinoma. J. Drug Delivery Sci. Technol. 2018;44:118–128. doi: 10.1016/j.jddst.2017.12.008. [DOI] [Google Scholar]
  436. Park C., Youn H., Kim H., Noh T., Kook Y. H., Oh E. T., Park H. J., Kim C.. Cyclodextrin-Covered Gold Nanoparticles for Targeted Delivery of an Anti-Cancer Drug. J. Mater. Chem. 2009;19(16):2310–2315. doi: 10.1039/b816209c. [DOI] [Google Scholar]
  437. Zhou H., Huang R., Su T., Li B., Zhou H., Ren J., Li Z.. A C-MWCNTs/AuNPs-Based Electrochemical Cytosensor to Evaluate the Anticancer Activity of Pinoresinol from Cinnamomum Camphora against HeLa Cells. Bioelectrochemistry. 2022;146:108133. doi: 10.1016/j.bioelechem.2022.108133. [DOI] [PubMed] [Google Scholar]
  438. Elangovan M., Santhoshkumar M., Selvaraj K., Sathishkumar K., Kumar M., Jothinathan M. K. D., Gatasheh M. K., Kumar Gaurav G., Rajesh K.. Sunlight-Driven Photocatalytic and Anticancer Properties of Biogenic Synthesized Gold Nanoparticles (AuNPs) Employing Polygala Elongata. J. King Saud Univ. Sci. 2024;36(5):103158. doi: 10.1016/j.jksus.2024.103158. [DOI] [Google Scholar]
  439. Babu B., Palanisamy S., Vinosha M., Anjali R., Kumar P., Pandi B., Tabarsa M., You S., Prabhu N. M.. Bioengineered Gold Nanoparticles from Marine Seaweed Acanthophora Spicifera for Pharmaceutical Uses: Antioxidant, Antibacterial, and Anticancer Activities. Bioprocess Biosyst. Eng. 2020;43(12):2231–2242. doi: 10.1007/s00449-020-02408-3. [DOI] [PubMed] [Google Scholar]
  440. Kumar D. S. R. S., Senthilkumar P., Surendran L., Sudhagar B.. Ganoderma lucidum-oriental mushroom mediated synthesis of gold nanoparticles conjugated with doxorubicin and evaluation of its anticancer potential on human breast cancer mcf-7/dox cells. Int. J. Pharm. Pharm. Sci. 2017;9(9):267–274. doi: 10.22159/ijpps.2017v9i9.20093. [DOI] [Google Scholar]
  441. Anjali Das C. G., Ganesh Kumar V., Dharani G., Stalin Dhas T., Karthick V., Vineeth Kumar C. M., Embrandiri A.. Macroalgae-Associated Halotolerant Marine Bacteria Exiguobacterium Aestuarii ADCG SIST3 Synthesized Gold Nanoparticles and Its Anticancer Activity in Breast Cancer Cell Line (MCF-7) J. Mol. Liq. 2023;383:122061. doi: 10.1016/j.molliq.2023.122061. [DOI] [Google Scholar]
  442. Alshahrani M. Y., Rafi Z., Alabdallah N. M., Shoaib A., Ahmad I., Asiri M., Zaman G. S., Wahab S., Saeed M., Khan S.. A Comparative Antibacterial, Antioxidant, and Antineoplastic Potential of Rauwolfia Serpentina (L.) Leaf Extract with Its Biologically Synthesized Gold Nanoparticles (R-AuNPs) Plants. 2021;10(11):2278. doi: 10.3390/plants10112278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  443. Venkatesh G., Serdaroğlu G., Üstün E., Haripriya D., Vennila P., Siva V., Haseena S., Sowmiya V., Pradhiksha A.. Green Synthesis, Characterization, Anti-Cancer and Antimicrobial Activity of AuNPs Extracted from Euphorbia Antiquorum Stem and Flower: Experimental and Theoretical Calculations. J. Drug Delivery Sci. Technol. 2024;95:105583. doi: 10.1016/j.jddst.2024.105583. [DOI] [Google Scholar]
  444. Musib D., Raza M. K., Pal M., Roy M. A.. Red Light-Activable MnI­(CO)­3-Functionalized Gold Nanocomposite as the Anticancer Prodrug with Theranostic Potential. Appl. Organomet. Chem. 2021;35(2):e6110. doi: 10.1002/aoc.6110. [DOI] [Google Scholar]
  445. Hosny M., Fawzy M., El-Badry Y. A., Hussein E. E., Eltaweil A. S.. Plant-Assisted Synthesis of Gold Nanoparticles for Photocatalytic, Anticancer, and Antioxidant Applications. J. Saudi Chem. Soc. 2022;26(2):101419. doi: 10.1016/j.jscs.2022.101419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  446. Oueslati M. H., Ben Tahar L., Harrath A. H.. Catalytic, Antioxidant and Anticancer Activities of Gold Nanoparticles Synthesized by Kaempferol Glucoside from Lotus Leguminosae. Arabian J. Chem. 2020;13(1):3112–3122. doi: 10.1016/j.arabjc.2018.09.003. [DOI] [Google Scholar]
  447. Ramalingam V., Revathidevi S., Shanmuganayagam T. S., Muthulakshmi L., Rajaram R.. Gold Nanoparticle Induces Mitochondria-Mediated Apoptosis and Cell Cycle Arrest in Nonsmall Cell Lung Cancer Cells. Gold Bull. 2017;50(2):177–189. doi: 10.1007/s13404-017-0208-x. [DOI] [Google Scholar]
  448. Abdelsattar A. S., Kamel A. G., Hussein A. H., Azzam M., Makky S., Rezk N., Essam K., Agwa M. M., El-Shibiny A.. The Promising Antibacterial and Anticancer Activity of Green Synthesized Zinc Nanoparticles in Combination with Silver and Gold Nanoparticles. J. Inorg. Organomet. Polym. Mater. 2023;33(7):1868–1881. doi: 10.1007/s10904-023-02614-y. [DOI] [Google Scholar]
  449. Seku K., Bhagavanth Reddy G., Osman A. I., Hussaini S. S., Kumar N. S., Al-Abri M., Pejjai B., Alreshaidan S. B., Al-Fatesh A. S., Kadimpati K. K.. Modified Frankincense Resin Stabilized Gold Nanoparticles for Enhanced Antioxidant and Synergetic Activity in In-Vitro Anticancer Studies. Int. J. Biol. Macromol. 2024;278:134935. doi: 10.1016/j.ijbiomac.2024.134935. [DOI] [PubMed] [Google Scholar]
  450. Gavade N. L., Deshmukh S. P., Dossou A. S., Raut S., Garadkar K. M.. Phytosynthesis of Anisotropic Silver and Gold Nanoparticles: Characterization and Anticancer Activity towards HIO180 and HeyA8 Cells. Spectrochim. Acta, Part A. 2025;333:125905. doi: 10.1016/j.saa.2025.125905. [DOI] [PubMed] [Google Scholar]
  451. Lomelí-Marroquín D., Cruz D. M., Nieto-Argüello A., Crua A. V., Chen J., Torres-Castro A., Webster T. J., Cholula-Díaz J. L.. Starch-Mediated Synthesis of Mono- and Bimetallic Silver/Gold Nanoparticles as Antimicrobial and Anticancer Agents. Int. J. Nanomed. 2019;14:2171–2190. doi: 10.2147/IJN.S192757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  452. Rohilla P., Chhikara A., Dalal M., Verma Y. K., Kumar S., Dahiya P.. Synthesis of Physalis Minima L. Based AuNPs: Insights in Their Antimicrobial, Antioxidant and Anticancer Activity. Inorg. Chem. Commun. 2025;178:114546. doi: 10.1016/j.inoche.2025.114546. [DOI] [Google Scholar]
  453. Akturk O.. The Anticancer Activity of Doxorubicin-Loaded Levan-Functionalized Gold Nanoparticles Synthesized by Laser Ablation. Int. J. Biol. Macromol. 2022;196:72–85. doi: 10.1016/j.ijbiomac.2021.12.030. [DOI] [PubMed] [Google Scholar]
  454. Li N., Chen Y., Zhang Y.-M., Yang Y., Su Y., Chen J.-T., Liu Y.. Polysaccharide-Gold Nanocluster Supramolecular Conjugates as a Versatile Platform for the Targeted Delivery of Anticancer Drugs. Sci. Rep. 2014;4(1):4164. doi: 10.1038/srep04164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  455. Mobaraki F., Momeni M., Taghavizadeh Yazdi M. E., Meshkat Z., Silanian Toosi M., Hosseini S. M.. Plant-Derived Synthesis and Characterization of Gold Nanoparticles: Investigation of Its Antioxidant and Anticancer Activity against Human Testicular Embryonic Carcinoma Stem Cells. Process Biochem. 2021;111:167–177. doi: 10.1016/j.procbio.2021.09.010. [DOI] [Google Scholar]
  456. Lee K. X., Shameli K., Nagao Y., Yew Y. P., Teow S.-Y., Moeini H.. Potential Use of Gold-Silver Core-Shell Nanoparticles Derived from Garcinia Mangostana Peel for Anticancer Compound, Protocatechuic Acid Delivery. Front. Mol. Biosci. 2022;9:997471. doi: 10.3389/fmolb.2022.997471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  457. Waghmare M. N., Qureshi T. S., Krishna C. M., Pansare K., Gadewal N., Hole A., Dongre P. M.. β-Lactoglobulin-Gold Nanoparticles Interface and Its Interaction with Some Anticancer Drugs – an Approach for Targeted Drug Delivery. J. Biomol. Struct. Dyn. 2022;40(13):6193–6210. doi: 10.1080/07391102.2021.1879270. [DOI] [PubMed] [Google Scholar]
  458. Punnoose M. S., Mathew B.. Microwave-Assisted Green Synthesis of Cyanthillium Cinereum Mediated Gold Nanoparticles: Evaluation of Its Antibacterial, Anticancer and Catalytic Degradation Efficacy. Res. Chem. Intermed. 2022;48(3):1025–1044. doi: 10.1007/s11164-021-04641-1. [DOI] [Google Scholar]
  459. Majumdar M., Biswas S. C., Choudhury R., Upadhyay P., Adhikary A., Roy D. N., Misra T. K.. Synthesis of Gold Nanoparticles Using Citrus Macroptera Fruit Extract: Anti-Biofilm and Anticancer Activity. ChemistrySelect. 2019;4(19):5714–5723. doi: 10.1002/slct.201804021. [DOI] [Google Scholar]
  460. Yang Z., Liu Z., Zhu J., Xu J., Pu Y., Bao Y.. Green Synthesis and Characterization of Gold Nanoparticles from Pholiota Adiposa and Their Anticancer Effects on Hepatic Carcinoma. Drug Delivery. 2022;29(1):997–1006. doi: 10.1080/10717544.2022.2056664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  461. Vairavel M., Devaraj E., Shanmugam R.. An Eco-Friendly Synthesis of Enterococcus Sp.–Mediated Gold Nanoparticle Induces Cytotoxicity in Human Colorectal Cancer Cells. Environ. Sci. Pollut. Res. 2020;27(8):8166–8175. doi: 10.1007/s11356-019-07511-x. [DOI] [PubMed] [Google Scholar]
  462. Sun B., Nanjun H., Leng H., Yanan P., Yu G., Chen K.. Anticancer Activity of Green Synthesised Gold Nanoparticles from Marsdenia Tenacissima Inhibits A549 Cell Proliferation through the Apoptotic Pathway. Artif. Cells Nanomed. Biotechnol. 2019;47(1):4012–4019. doi: 10.1080/21691401.2019.1575844. [DOI] [PubMed] [Google Scholar]
  463. Ke Y., Aboody M. S. Al., Alturaiki W., Alsagaby S. A., Alfaiz F. A., Veeraraghavan V. P., Mickymaray S.. Photosynthesized Gold Nanoparticles from Catharanthus Roseus Induces Caspase-Mediated Apoptosis in Cervical Cancer Cells (HeLa) Artif. Cells Nanomed. Biotechnol. 2019;47(1):1938–1946. doi: 10.1080/21691401.2019.1614017. [DOI] [PubMed] [Google Scholar]
  464. Dhandapani S., Xu X., Wang R., Puja A. M., Kim H., Perumalsamy H., Balusamy S. R., Kim Y.-J.. Biosynthesis of Gold Nanoparticles Using Nigella Sativa and Curtobacterium Proimmune K3 and Evaluation of Their Anticancer Activity. Mater. Sci. Eng.: C. 2021;127:112214. doi: 10.1016/j.msec.2021.112214. [DOI] [PubMed] [Google Scholar]
  465. Patil M. P., Jin X., Simeon N. C., Palma J., Kim D., Ngabire D., Kim N.-H., Tarte N. H., Kim G.-D.. Anticancer Activity of Sasa Borealis Leaf Extract-Mediated Gold Nanoparticles. Artif. Cells Nanomed. Biotechnol. 2018;46(1):82–88. doi: 10.1080/21691401.2017.1293675. [DOI] [PubMed] [Google Scholar]
  466. Jayaseelan C., Upadhyay P., Sahal D., Kamaraj C., Thirugnanasambandam R., Siva D., Saravanan D., Regina Mary R.. Biosynthesis of Gold Nanoparticles Mediated by Medicinal Phytometabolites: An Effective Tool against Plasmodium Falciparum and Human Breast Cancer Cells. J. Drug Delivery Sci. Technol. 2024;95:105520. doi: 10.1016/j.jddst.2024.105520. [DOI] [Google Scholar]
  467. Koc-Bilican, B. ; Benarous, S. ; Koca, F. D. ; Cansaran-Duman, D. ; Sørensen, M. V. ; Bilican, I. ; Kaya, M. . Anti-Cancer Effect of Thymus Vulgaris Based Synthesized Gold Nanoparticles in Giant Macroporous Silica: Impact on MCF-7 Breast Cancer Cells J. Aust. Ceram. Soc. 2025. 10.1007/s41779-025-01164-5. [DOI]
  468. El-Naggar N. E.-A., Rabei N. H., Elmansy M. F., Elmessiry O. T., El-Sherbeny M. K., El-Saidy M. E., Sarhan M. T., Helal M. G.. Artificial Neural Network Approach for Prediction of AuNPs Biosynthesis by Streptomyces Flavolimosus, Characterization, Antitumor Potency in-Vitro and in-Vivo against Ehrlich Ascites Carcinoma. Sci. Rep. 2023;13(1):12686. doi: 10.1038/s41598-023-39177-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  469. Sharma M., Monika, Thakur P., Saini R. V., Kumar R., Torino E.. Unveiling Antimicrobial and Anticancerous Behavior of AuNPs and AgNPs Moderated by Rhizome Extracts of Curcuma Longa from Diverse Altitudes of Himalaya. Sci. Rep. 2020;10(1):10934. doi: 10.1038/s41598-020-67673-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  470. Mary S. J., Veeravarmal V., Thankappan P., Arumugam P., Augustine P. I., Franklin R.. Anti-Cancer Effects of Green Synthesized Gold Nanoparticles Using Leaf Extract of Annona Muricata. L against Squamous Cell Carcinoma Cell Line 15 through Apoptotic Pathway. Dent. Res. J. 2024;21(1):14. doi: 10.4103/drj.drj_521_23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  471. Gawali P., Ramteke L., Jadhav B. L., Khade B. S.. Trypsin Conjugated Au Nanoparticles Using Sonneratia Alba Fruits: Interaction and Binding Studies with Antioxidant, Anti-Inflammatory, and Anticancer Activities. J. Cluster Sci. 2023;34(6):2759–2779. doi: 10.1007/s10876-023-02416-9. [DOI] [Google Scholar]
  472. Namasivayam S. K. R., Haveela E., Samrat K., Bharani R. S. A., Sigamani S., Srinivasan R., Kavisri M., Moovendhan M.. Production, Nanoformulation, and Potential Anticancer Effect of Camptothecin Metabolites from Biomass of Aspergillus Niger. Biomass Convers. Biorefin. 2024;14(15):18005–18018. doi: 10.1007/s13399-023-03861-2. [DOI] [Google Scholar]
  473. Wu T., Duan X., Hu C., Wu C., Chen X., Huang J., Liu J., Cui S.. Synthesis and Characterization of Gold Nanoparticles from Abies Spectabilis Extract and Its Anticancer Activity on Bladder Cancer T24 Cells. Artif. Cells Nanomed. Biotechnol. 2019;47(1):512–523. doi: 10.1080/21691401.2018.1560305. [DOI] [PubMed] [Google Scholar]
  474. Ayyanaar S., Kesavan M. P.. One-Pot Biogenic Synthesis of Gold Nanoparticles@saponins Niosomes: Sustainable Nanomedicine for Antibacterial, Anti-Inflammatory and Anticancer Therapeutics. Colloids Surf., A. 2023;676:132229. doi: 10.1016/j.colsurfa.2023.132229. [DOI] [Google Scholar]
  475. Sekar V., Santhanam A., Arunkumar P.. PH-Responsive Gold Nanoparticle/PVP Nanoconjugate for Targeted Delivery and Enhanced Anticancer Activity of Withaferin A. Processes. 2025;13(5):1290. doi: 10.3390/pr13051290. [DOI] [Google Scholar]
  476. Shunmugam R., Balusamy S. R., Kumar V., Menon S., Lakshmi T., Perumalsamy H.. Biosynthesis of Gold Nanoparticles Using Marine Microbe (Vibrio Alginolyticus) and Its Anticancer and Antioxidant Analysis. J. King Saud Univ. Sci. 2021;33(1):101260. doi: 10.1016/j.jksus.2020.101260. [DOI] [Google Scholar]
  477. Shinde A. S., Mendhulkar V. D.. Anticancer Activity of Gold Nanobioconjugates Synthesized from Elephantopus Scaber (Linn.) Leaf Extract. J. Cancer Res. Ther. 2023;19(Suppl 1):S250–S259. doi: 10.4103/jcrt.JCRT_1043_20. [DOI] [PubMed] [Google Scholar]
  478. Leng M., Jiang H., Zhang S., Bao Y.. Green Synthesis of Gold Nanoparticles from Polygahatous Polysaccharides and Their Anticancer Effect on Hepatic Carcinoma through Immunoregulation. ACS Omega. 2024;9(19):21144–21151. doi: 10.1021/acsomega.4c01025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  479. Vinay S. P., Udayabhanu, Sumedha H. N., Shashank M., Nagaraju G., Chandrasekhar N.. In-Vitro Antibacterial, Antioxidant and Cytotoxic Potential of Gold Nanoparticles Synthesized Using Novel Elaeocarpus Ganitrus Seeds Extract. J. Sci.: Adv. Mater. Devices. 2021;6(1):127–133. doi: 10.1016/j.jsamd.2020.09.008. [DOI] [Google Scholar]
  480. Ielo I., Rando G., Giacobello F., Sfameni S., Castellano A., Galletta M., Drommi D., Rosace G., Plutino M. R.. Synthesis, Chemical–Physical Characterization, and Biomedical Applications of Functional Gold Nanoparticles: A Review. Molecules. 2021;26(19):5823. doi: 10.3390/molecules26195823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  481. Anik M. I., Mahmud N., Al Masud A., Hasan M.. Gold Nanoparticles (GNPs) in Biomedical and Clinical Applications: A Review. Nano Select. 2022;3(4):792–828. doi: 10.1002/nano.202100255. [DOI] [Google Scholar]
  482. Gowtham P., Girigoswami K., Prabhu A. D., Pallavi P., Thirumalai A., Harini K., Girigoswami A.. Hydrogels of Alginate Derivative-Encased Nanodots Featuring Carbon-Coated Manganese Ferrite Cores with Gold Shells to Offer Antiangiogenesis with Multimodal Imaging-Based Theranostics. Adv. Ther. 2024;7(6):2400054. doi: 10.1002/adtp.202400054. [DOI] [Google Scholar]
  483. Aydogan B., Li J., Rajh T., Chaudhary A., Chmura S. J., Pelizzari C., Wietholt C., Kurtoglu M., Redmond P.. AuNP-DG: Deoxyglucose-Labeled Gold Nanoparticles as X-Ray Computed Tomography Contrast Agents for Cancer Imaging. Mol. Imaging Biol. 2010;12(5):463–467. doi: 10.1007/s11307-010-0299-8. [DOI] [PubMed] [Google Scholar]
  484. Nguyen V. P., Qian W., Li Y., Liu B., Aaberg M., Henry J., Zhang W., Wang X., Paulus Y. M.. Chain-like Gold Nanoparticle Clusters for Multimodal Photoacoustic Microscopy and Optical Coherence Tomography Enhanced Molecular Imaging. Nat. Commun. 2021;12(1):34. doi: 10.1038/s41467-020-20276-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  485. Queiroz S. M., Veriato T. S., Raniero L., Castilho M. L.. Gold Nanoparticles Conjugated with Epidermal Growth Factor and Gadolinium for Precision Delivery of Contrast Agents in Magnetic Resonance Imaging. Radiol. Phys. Technol. 2024;17(1):153–164. doi: 10.1007/s12194-023-00761-y. [DOI] [PubMed] [Google Scholar]
  486. Zeng X., Tang L., Zhang W., Hong X., Xiao Y.. Shape and Size Effects of Gold Nanoparticles for Tumor Photoacoustic Imaging and Photothermal Therapy Within the NIR-I and NIR-II Biowindows. Small. 2025;21:2412296. doi: 10.1002/smll.202412296. [DOI] [PubMed] [Google Scholar]
  487. Lansangan C., Khoobchandani M., Jain R., Rudensky S., Perry C. C., Patil R.. Designing Gold Nanoparticles for Precise Glioma Treatment: Challenges and Alternatives. Materials. 2024;17:1153. doi: 10.3390/ma17051153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  488. Truong T. T., Mondal S., Doan V. H. M., Tak S., Choi J., Oh H., Nguyen T. D., Misra M., Lee B., Oh J.. Precision-Engineered Metal and Metal-Oxide Nanoparticles for Biomedical Imaging and Healthcare Applications. Adv. Colloid Interface Sci. 2024;332:103263. doi: 10.1016/j.cis.2024.103263. [DOI] [PubMed] [Google Scholar]
  489. Abrishami A., Bahrami A. R., Nekooei S., Saljooghi A. S., Matin M. M.. Hybridized Quantum Dot, Silica, and Gold Nanoparticles for Targeted Chemo-Radiotherapy in Colorectal Cancer Theranostics. Commun. Biol. 2024;7(1):393. doi: 10.1038/s42003-024-06043-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  490. Zhang R., Thoröe-Boveleth S., Chigrin D. N., Kiessling F., Lammers T., Pallares R. M.. Nanoscale Engineering of Gold Nanostars for Enhanced Photoacoustic Imaging. J. Nanobiotechnol. 2024;22(1):115. doi: 10.1186/s12951-024-02379-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  491. Dai X., Zhao X., Liu Y., Chen B., Ding X., Zhao N., Xu F. J.. Controlled Synthesis and Surface Engineering of Janus Chitosan-Gold Nanoparticles for Photoacoustic Imaging-Guided Synergistic Gene/Photothermal Therapy. Small. 2021;17(11):2006004. doi: 10.1002/smll.202006004. [DOI] [PubMed] [Google Scholar]
  492. Lv Q., Zhang Y., Yang R., Dai Y., Lin Y., Sun K., Xu H., Tao K.. Photoacoustic Imaging Endometriosis Lesions with Nanoparticulate Polydopamine as a Contrast Agent. Adv. Healthcare Mater. 2024;13(2):2302175. doi: 10.1002/adhm.202302175. [DOI] [PubMed] [Google Scholar]
  493. Wang W., Wang J., Ding Y.. Gold Nanoparticle-Conjugated Nanomedicine: Design, Construction, and Structure-Efficacy Relationship Studies. J. Mater. Chem. B. 2020;8:4813–4830. doi: 10.1039/c9tb02924a. [DOI] [PubMed] [Google Scholar]
  494. Zazo H., Colino C. I., Gutiérrez-Millán C., Cordero A. A., Bartneck M., Lanao J. M.. Physiologically Based Pharmacokinetic (PBPK) Model of Gold Nanoparticle-Based Drug Delivery System for Stavudine Biodistribution. Pharmaceutics. 2022;14(2):406. doi: 10.3390/pharmaceutics14020406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  495. Sun C., Tian Y., Li Z., Zhao L., Ji T., Lu Y.. Hexanoyl Glycol Chitosan/Gold Nanoparticle-Based Thermosensitive Nanoagent for Synergistic Photothermal/Chemotherapy of Tumors. ACS Appl. Nano Mater. 2024;7(15):17659–17667. doi: 10.1021/acsanm.4c02939. [DOI] [Google Scholar]
  496. Devi N., Singh P., Sharma R., Kumar M., Pandey S. K., Sharma R. K., Wangoo N.. A Lysine-Rich Cell Penetrating Peptide Engineered Multifunctional Gold Nanoparticle-Based Drug Delivery System with Enhanced Cellular Penetration and Stability. J. Mater. Sci. 2022;57(35):16842–16857. doi: 10.1007/s10853-022-07681-z. [DOI] [Google Scholar]
  497. Zheng K., Zhou D., Wu L., Li J., Zhao B., Zhang S., He R., Xiao L., Zoya I., Yu L., Zhang Y., Li Y., Gao J., Li K.. K. ZHENG ET AL.Gold-Nanoparticle-Based Multistage Drug Delivery System for Antitumor Therapy. Drug Delivery. 2022;29(1):3186–3196. doi: 10.1080/10717544.2022.2128469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  498. Amalkar S. V., Hatwar P. R., Bakal R. L., Kohale N. B.. Advance in Gold Nanoparticle- Mediated Drug Delivery System. GSC Biol. Pharm. Sci. 2024;28(3):169–179. doi: 10.30574/gscbps.2024.28.3.0324. [DOI] [Google Scholar]
  499. Georgeous J., AlSawaftah N., Abuwatfa W. H., Husseini G. A.. Review of Gold Nanoparticles: Synthesis, Properties, Shapes, Cellular Uptake, Targeting, Release Mechanisms and Applications in Drug Delivery and Therapy. Pharmaceutics. 2024;16:1332. doi: 10.3390/pharmaceutics16101332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  500. Feng Z. V., Gunsolus I. L., Qiu T. A., Hurley K. R., Nyberg L. H., Frew H., Johnson K. P., Vartanian A. M., Jacob L. M., Lohse S. E., Torelli M. D., Hamers R. J., Murphy C. J., Haynes C. L.. Impacts of Gold Nanoparticle Charge and Ligand Type on Surface Binding and Toxicity to Gram-Negative and Gram-Positive Bacteria. Chem. Sci. 2015;6(9):5186–5196. doi: 10.1039/C5SC00792E. [DOI] [PMC free article] [PubMed] [Google Scholar]
  501. Li Q., Ma C., Ma Y., Ma Y., Mao Y., Meng Z.. Sustained Bimatoprost Release Using Gold Nanoparticles Laden Contact Lenses. J. Biomater. Sci. Polym. Ed. 2021;32(12):1618–1634. doi: 10.1080/09205063.2021.1927656. [DOI] [PubMed] [Google Scholar]
  502. Dykman L., Khlebtsov B., Khlebtsov N.. Drug Delivery Using Gold Nanoparticles. Adv. Drug Delivery Rev. 2025;216:115481. doi: 10.1016/j.addr.2024.115481. [DOI] [PubMed] [Google Scholar]
  503. Nichols J. W., Bae Y. H.. EPR: Evidence and Fallacy. J. Controlled Release. 2014;190:451–464. doi: 10.1016/j.jconrel.2014.03.057. [DOI] [PubMed] [Google Scholar]
  504. Sengupta A., Bhattacharya D., Lahiri D., Nag M., Dey A., Ray S.. Gold and Silver Nanoparticles as Potent Quorum Quenchers: A Critical Review. Bionanoscience. 2025;15(1):73. doi: 10.1007/s12668-024-01721-5. [DOI] [Google Scholar]
  505. Elshaer S. L., Shaaban M. I.. Inhibition of Quorum Sensing and Virulence Factors of Pseudomonas Aeruginosa by Biologically Synthesized Gold and Selenium Nanoparticles. Antibiotics. 2021;10(12):1461. doi: 10.3390/antibiotics10121461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  506. Qais F. A., Ahmad I., Altaf M., Alotaibi S. H.. Biofabrication of Gold Nanoparticles Using Capsicum Annuum Extract and Its Antiquorum Sensing and Antibiofilm Activity against Bacterial Pathogens. ACS Omega. 2021;6:16670–16682. doi: 10.1021/acsomega.1c02297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  507. Singh R. P., Ramarao P.. Cellular Uptake, Intracellular Trafficking and Cytotoxicity of Silver Nanoparticles. Toxicol. Lett. 2012;213(2):249. doi: 10.1016/j.toxlet.2012.07.009. [DOI] [PubMed] [Google Scholar]
  508. Ahmed S. A. K. S., Rudden M., Smyth T. J., Dooley J. S. G., Marchant R., Banat I. M.. Natural Quorum Sensing Inhibitors Effectively Downregulate Gene Expression of Pseudomonas Aeruginosa Virulence Factors. Appl. Microbiol. Biotechnol. 2019;103(8):3521–3535. doi: 10.1007/s00253-019-09618-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  509. Khosravi M., Mirzaie A., Kashtali A. B., Noorbazargan H.. Antibacterial, Anti-Efflux, Anti-Biofilm, Anti-Slime (Exopolysaccharide) Production and Urease Inhibitory Efficacies of Novel Synthesized Gold Nanoparticles Coated Anthemis Atropatana Extract against Multidrug- Resistant Klebsiella Pneumoniae Strains. Arch. Microbiol. 2020;202:2105–2115. doi: 10.1007/s00203-020-01930-y. [DOI] [PubMed] [Google Scholar]
  510. Chahande A. M., Lathigara D., Prabhune A. A., Devi R. N.. Red Fluorescent Ultra-Small Gold Nanoclusters Functionalized with Signal Molecules to Probe Specificity in Quorum Sensing Receptors in Gram-Negative Bacteria. Arch. Microbiol. 2021;203(7):4293–4301. doi: 10.1007/s00203-021-02338-y. [DOI] [PubMed] [Google Scholar]
  511. Zhang Y., Li P., Su R., Wen F., Jia Z., Lv Y., Cai J., Su W.. Curcumin-Loaded Multifunctional Chitosan Gold Nanoparticles: An Enhanced PDT/PTT Dual-Modal Phototherapeutic and PH-Responsive Antimicrobial Agent. Photodiagn. Photodyn. Ther. 2022;39:103011. doi: 10.1016/j.pdpdt.2022.103011. [DOI] [PubMed] [Google Scholar]
  512. Vinoj G., Rashmirekha P., Avinash S., Baskaralingam V.. In Vitro Cytotoxic Effects of Gold Nanoparticles Coated with Functional Acyl Homoserine Lactone Lactonase Protein from Bacillus Licheniformis and Their Antibiofilm Activity against Proteus Species. Antimicrob. Agents Chemother. 2015;59(2):763–771. doi: 10.1128/aac.03047-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  513. Tabassum N., Jeong G.-J., Jo D.-M., Khan F., Kim Y.-M.. Attenuation of Biofilm and Virulence Factors of Pseudomonas Aeruginosa by Tetramethylpyrazine-Gold Nanoparticles. Microb. Pathog. 2024;191:106658. doi: 10.1016/j.micpath.2024.106658. [DOI] [PubMed] [Google Scholar]
  514. Govindasamy B.. Multifaceted Toxicity Assessment of Au, Ag, and TiO2 Nanoparticles Synthesized by Quorum Quenching Bacterium Salmonella Bongori: Impact on Bacterial Pathogens, Cancer Cells, Mosquitoes, Zebrafish, and Brine Shrimp. Process Saf. Environ. Prot. 2024;189:960–976. doi: 10.1016/j.psep.2024.06.099. [DOI] [Google Scholar]
  515. Fernando S. I. D., Cruz K. G. J.. Ethnobotanical Biosynthesis of Gold Nanoparticles and Its Downregulation of Quorum Sensing-Linked AhyR Gene in Aeromonas Hydrophila. SN Appl. Sci. 2020;2(4):570. doi: 10.1007/s42452-020-2368-1. [DOI] [Google Scholar]
  516. Namasivayam S. K. R., Venkatachalam G., Bharani R. S. A.. Immuno Biocompatibility and Anti-Quorum Sensing Activities of Chitosan-Gum Acacia Gold Nanocomposite (CS-GA-AuNC) against Pseudomonas Aeruginosa Drug-Resistant Pathogen. Sustainable Chem. Pharm. 2020;17:100300. doi: 10.1016/j.scp.2020.100300. [DOI] [Google Scholar]
  517. Jayeoye T. J., Nwude E. F., Singh S., Prajapati B. G., Kapoor D. U., Muangsin N.. Sustainable Synthesis of Gold Nanoparticles for Drug Delivery and Cosmeceutical Applications: A Review. Bionanoscience. 2024;14(3):3355–3384. doi: 10.1007/s12668-024-01436-7. [DOI] [Google Scholar]
  518. Ranade T., Sati A., Pratap A., Mali S. N.. Curcumin-Integrated Biopolymer Films for Active Packaging: Current Trends and Future Directions. Chem. Pap. 2025;79:1303–1334. doi: 10.1007/s11696-025-03892-1. [DOI] [Google Scholar]
  519. Mali S. N., Pandey A.. Development of Curcumin Integrated Smart PH Indicator, Antibacterial, and Antioxidant Waste Derived Artocarpus Lakoocha Starch-Based Packaging Film. Int. J. Biol. Macromol. 2024;275:133827. doi: 10.1016/j.ijbiomac.2024.133827. [DOI] [PubMed] [Google Scholar]
  520. Abidin N. A. Z., Kormin F., Abidin N. A. Z., Bakar M. F. A., Moujdin I. A.. Synthesis and Characterization of Curcumin-Chitosan Loaded Gold Nanoparticles by Oryctes Rhinoceros’ Chitin for Cosmeceutical Application. Molecules. 2023;28(4):1799. doi: 10.3390/molecules28041799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  521. Amir S. N. K. b. M., Nordin M. F. b. M., Shameli K., Hamid M. A.. Gold Nanoparticles from Plant Materials: Green Extraction, Biological Synthesis and Its Beneficials Properties for Cosmeceutical Applications. J. Res. Nanosci. Nanotechnol. 2021;2(1):12–29. doi: 10.37934/jrnn.2.1.1229. [DOI] [Google Scholar]
  522. Cardoza C., Nagtode V., Pratap A., Mali S. N.. Emerging Applications of Nanotechnology in Cosmeceutical Health Science: Latest Updates. Health Sci. Rev. 2022;4:100051. doi: 10.1016/j.hsr.2022.100051. [DOI] [Google Scholar]
  523. Rudolf R., Majerič P., Jelen Z., Horvat A., Krajnc D.. The Environmental Impact of Using Gold Nanoparticles and 3HFWC in Cosmetics, as Determined with LCA Methodology. Rev. Inorg. Chem. 2024;44:409–419. doi: 10.1515/revic-2024-0012. [DOI] [Google Scholar]
  524. Singpanna K., Pornpitchanarong C., Patrojanasophon P., Rojanarata T., Ngawhirunpat T., Li S. K., Opanasopit P.. Chitosan Capped-Gold Nanoparticles as Skin Penetration Enhancer for Small Molecules: A Study in Porcine Skin. Int. J. Pharm. 2023;640:123034. doi: 10.1016/j.ijpharm.2023.123034. [DOI] [PubMed] [Google Scholar]
  525. Singh S., Chunglok W., Nwabor O. F., Ushir Y. V., Singh S., Panpipat W.. Hydrophilic Biopolymer Matrix Antibacterial Peel-off Facial Mask Functionalized with Biogenic Nanostructured Material for Cosmeceutical Applications. J. Polym. Environ. 2022;30(3):938–953. doi: 10.1007/s10924-021-02249-5. [DOI] [Google Scholar]
  526. Kim H. M., Park J. H., Choi Y. J., Oh J. M., Park J.. Hyaluronic Acid-Coated Gold Nanoparticles as a Controlled Drug Delivery System for Poorly Water-Soluble Drugs. RSC Adv. 2023;13(8):5529–5537. doi: 10.1039/D2RA07276A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  527. Permatasari A. D., Syukri Y., Werdyani S.. Gold Nanoparticles with Natural Ingredients as Anti-Aging: A Systematic Review. J. Pharm. Sci. Commun. 2023;20(1):90–113. doi: 10.24071/jpsc.004374. [DOI] [Google Scholar]
  528. Senthilkumar P., Surendran L., Sudhagar B., Kumar D. S. R. S.. Facile Green Synthesis of Gold Nanoparticles from Marine Algae Gelidiella Acerosa and Evaluation of Its Biological Potential. SN Appl. Sci. 2019;1(4):284. doi: 10.1007/s42452-019-0284-z. [DOI] [Google Scholar]
  529. Ben Haddada M., Gerometta E., Chawech R., Sorres J., Bialecki A., Pesnel S., Spadavecchia J., Morel A.-L.. Assessment of Antioxidant and Dermoprotective Activities of Gold Nanoparticles as Safe Cosmetic Ingredient. Colloids Surf., B. 2020;189:110855. doi: 10.1016/j.colsurfb.2020.110855. [DOI] [PubMed] [Google Scholar]
  530. Gubitosa J., Rizzi V., Lopedota A., Fini P., Laurenzana A., Fibbi G., Fanelli F., Petrella A., Laquintana V., Denora N., Comparelli R., Cosma P.. One Pot Environmental Friendly Synthesis of Gold Nanoparticles Using Punica Granatum Juice: A Novel Antioxidant Agent for Future Dermatological and Cosmetic Applications. J. Colloid Interface Sci. 2018;521:50–61. doi: 10.1016/j.jcis.2018.02.069. [DOI] [PubMed] [Google Scholar]
  531. Abidin N. A. Z., Kormin F., Abidin N. A. Z., Bakar M. F. A., Moujdin I. A.. Synthesis and Characterization of Curcumin-Chitosan Loaded Gold Nanoparticles by Oryctes Rhinoceros’ Chitin for Cosmeceutical Application. Molecules. 2023;28(4):1799. doi: 10.3390/molecules28041799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  532. Jiménez-Pérez Z. E., Singh P., Kim Y.-J., Mathiyalagan R., Kim D.-H., Lee M. H., Yang D. C.. Applications of Panax Ginseng Leaves-Mediated Gold Nanoparticles in Cosmetics Relation to Antioxidant, Moisture Retention, and Whitening Effect on B16BL6 Cells. J. Ginseng Res. 2018;42(3):327–333. doi: 10.1016/j.jgr.2017.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  533. De Canha M. N., Thipe V. C., Katti K. V., Mandiwana V., Kalombo M. L., Ray S. S., Rikhotso R., Janse van Vuuren A., Lall N.. The Activity of Gold Nanoparticles Synthesized Using Helichrysum Odoratissimum Against Cutibacterium Acnes Biofilms. Front. Cell Dev. Biol. 2021;9:675064. doi: 10.3389/fcell.2021.675064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  534. Silveira P. C. L., Rodrigues M. S., Gelain D. P., de Oliveira J.. Gold Nanoparticles Application to the Treatment of Brain Dysfunctions Related to Metabolic Diseases: Evidence from Experimental Studies. Metab. Brain Dis. 2023;38(1):123–135. doi: 10.1007/s11011-022-00929-2. [DOI] [PubMed] [Google Scholar]
  535. Chiang M.-C., Yang Y.-P., Nicol C. J. B., Wang C.-J.. Gold Nanoparticles in Neurological Diseases: A Review of Neuroprotection. Int. J. Mol. Sci. 2024;25(4):2360. doi: 10.3390/ijms25042360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  536. Hussein J., El-Naggar M. E., Fouda M. M. G., Othman S. I., Allam A. A., Nadwa E. H., Rashwan E. K., Hendawy O. M.. Eco-Friendly Microwave Synthesis of Gold Nanoparticles for Attenuation of Brain Dysfunction in Diabetic Rats. J. Cluster Sci. 2021;32(2):423–435. doi: 10.1007/s10876-020-01801-y. [DOI] [Google Scholar]
  537. Prá M., Ferreira G. K., de Mello A. H., Uberti M. F., Engel N. A., Costa A. B., Zepon K. M., Francisco G. G., Hlavac N. R. C., Terra S. R., Garcez M. L., Zaccaron R. P., Mendes C., Tschoeke A. C. P., Kanis L. A., Budni J., Silveira P. C. L., Petronilho F., da Silva Paula M. M., Rezin G. T.. Treatment with Isolated Gold Nanoparticles Reverses Brain Damage Caused by Obesity. Mater. Sci. Eng.: C. 2021;120:111392. doi: 10.1016/j.msec.2020.111392. [DOI] [PubMed] [Google Scholar]
  538. Wu A. L., Wu A. F., Chen C. Y., Moreno R. L. H., Wu J. L., Wong P. C.. Laser-Induced Photothermal Hydrogels Promote the Proliferation of MC3T3-E1 Preosteoblasts for Enhanced Bone Healing. J. Funct. Biomater. 2025;16(2):63. doi: 10.3390/jfb16020063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  539. Badir A., Refki S., Sekkat Z.. Utilizing Gold Nanoparticles in Plasmonic Photothermal Therapy for Cancer Treatment. Heliyon. 2025:e42738. doi: 10.1016/j.heliyon.2025.e42738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  540. Faid A. H., Hussein F. E. Z., Mostafa E. M., Shouman S. A., Badr Y. A., Sliem M. A.. Hybrid Chitosan Gold Nanoparticles for Photothermal Therapy and Enhanced Cytotoxic Action of 6-Mercaptopurine on Breast Cancer Cell Line. Beni Suef Univ J. Basic Appl. Sci. 2023;12(1):83. doi: 10.1186/s43088-023-00419-z. [DOI] [Google Scholar]
  541. Frantellizzi V., Verrina V., Raso C., Pontico M., Petronella F., Bertana V., Ballesio A., Marasso S. L., Miglietta S., Rosa P., Scibetta S., Petrozza V., De Feo M. S., De Vincentis G., Calogero A., Pani R., Perotto G., De Sio L.. 99mTc-Labeled Keratin Gold-Nanoparticles in a Nephron-like Microfluidic Chip for Photo-Thermal Therapy Applications. Mater. Today Adv. 2022;16:100286. doi: 10.1016/j.mtadv.2022.100286. [DOI] [Google Scholar]
  542. Darvish S., Budala D. G., Goriuc A.. Antibacterial Properties of an Experimental Dental Resin Loaded with Gold Nanoshells for Photothermal Therapy Applications. J. Funct. Biomater. 2024;15(4):100. doi: 10.3390/jfb15040100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  543. Vashisth P., Smith C. L., Amarasekara D. L., Dasanyake G. S., Singh G., Chism C. M., Hamadani C. M., Shaikh T., Grovich N., Gamboa B., Fitzkee N. C., Hammer N. I., Tanner E. E. L.. Choline Carboxylic Acid Ionic Liquid-Stabilized Anisotropic Gold Nanoparticles for Photothermal Therapy. ACS Appl. Nano Mater. 2023;7:26332–26343. doi: 10.1021/acsanm.3c04645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  544. Tian Y., Jia D., Dirican M., Cui M., Fang D., Yan C., Xie J., Liu Y., Li C., Fu J., Liu H., Chen G., Zhang X., Tao J.. Highly Soluble and Stable, High Release Rate Nanocellulose Codrug Delivery System of Curcumin and AuNPs for Dual Chemo-Photothermal Therapy. Biomacromolecules. 2022;23(3):960–971. doi: 10.1021/acs.biomac.1c01367. [DOI] [PubMed] [Google Scholar]
  545. Aliyah N., Nasution M. A. F., Ayudia Putri Y. M. T., Gunlazuardi J., Ivandini T. A.. Modification of Carbon Foam with 4-Mercaptobenzoic Acid Functionalised Gold Nanoparticles for an Application in a Yeast-Based Microbial Fuel Cell. RSC Adv. 2022;12(44):28647–28657. doi: 10.1039/D2RA05100A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  546. Zhu G., Sun Z., Hui P., Chen W., Jiang X.. Composite Film with Antibacterial Gold Nanoparticles and Silk Fibroin for Treating Multidrug-Resistant E. Coli-Infected Wounds. ACS Biomater. Sci. Eng. 2021;7(5):1827–1835. doi: 10.1021/acsbiomaterials.0c01271. [DOI] [PubMed] [Google Scholar]
  547. Piktel E., Suprewicz Ł., Depciuch J., Chmielewska S., Skłodowski K., Daniluk T., Król G., Kołat-Brodecka P., Bijak P., Pajor-Świerzy A., Fiedoruk K., Parlinska-Wojtan M., Bucki R.. Varied-Shaped Gold Nanoparticles with Nanogram Killing Efficiency as Potential Antimicrobial Surface Coatings for the Medical Devices. Sci. Rep. 2021;11(1):12546. doi: 10.1038/s41598-021-91847-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  548. Wang Y., Wu F., Li Y., Wang S., Ren Y., Shi L., van der Mei H. C., Liu Y.. Ellagic Acid-Modified Gold Nanoparticles to Combat Multi-Drug Resistant Bacterial Infections in Vitro and in Vivo. Mater. Horiz. 2024;11(19):4781–4790. doi: 10.1039/D4MH00642A. [DOI] [PubMed] [Google Scholar]
  549. Wang L., Zheng W., Li S., Zhong L., Jiang X.. Aminophenol-Decorated Gold Nanoparticles for Curing Bacterial Infections. Nano Lett. 2022;22(9):3576–3582. doi: 10.1021/acs.nanolett.1c04968. [DOI] [PubMed] [Google Scholar]
  550. Wang L., Zheng W., Li S., Hou Q., Jiang X.. Modulating the Antibacterial Activity of Gold Nanoparticles by Balancing Their Monodispersity and Aggregation. Chem. Commun. 2022;58(55):7690–7693. doi: 10.1039/D2CC02782H. [DOI] [PubMed] [Google Scholar]
  551. Ndugire W., Truong D., Raviranga N. G. H., Lao J., Ramström O., Yan M.. Turning on the Antimicrobial Activity of Gold Nanoclusters Against Multidrug-Resistant Bacteria. Angew. Chem., Int. Ed. 2023;62(11):e202214086. doi: 10.1002/anie.202214086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  552. Shalini A., Priya K., Kothai S., Pandian K., Anbalagan G., Jaisankar V.. Synthesis and Characterisation of Graphene Oxide Decorated Gold Nano Particles and Their Application towards Antibacterial Activity. Chem. Pap. 2022;76(11):6861–6867. doi: 10.1007/s11696-022-02375-x. [DOI] [Google Scholar]
  553. Shanwaz Md. M., Shyam P.. Anti-Bacterial Effect and Characteristics of Gold Nanoparticles (AuNps) Formed with Vitex Negundo Plant Extract. Appl. Biochem. Biotechnol. 2023;195(3):1630–1643. doi: 10.1007/s12010-022-04217-8. [DOI] [PubMed] [Google Scholar]
  554. de Resende Marques R., Oliveira N. A., Rodriguez S. R., Alvarez L. A. C., Silva L. P. C., de Oliveira J. P., Endringer D. C., Schuenck R. P., Guimarães M. C. C.. In Vitro Antibacterial Effects of Gold Nanoparticles Synthesized with Virola Oleifera on Staphylococcus Aureus and Pseudomonas Aeruginosa Strains. Bionanoscience. 2023;13(3):900–910. doi: 10.1007/s12668-023-01152-8. [DOI] [Google Scholar]
  555. Hamedani H., Kariminezhad H., Amani H.. Photothermal Antibacterial Activity of Gold Nanorods Stabilized by Phospholipid. Braz. J.Phys. 2023;53(2):50. doi: 10.1007/s13538-023-01265-1. [DOI] [Google Scholar]
  556. Deivanathan S. K., Prakash J. T. J., Bhagyalakshmi J., Rosaiah P., Reddy I. N., Bai C., Karim M. R.. Bio-Fabrication of Gold Nanoparticles Using Codariocalyx Motorius for Antimicrobial, Antioxidant and Anticancer Activity. Waste Biomass Valorization. 2025;16:1927–1938. doi: 10.1007/s12649-024-02778-9. [DOI] [Google Scholar]
  557. Aksoy A., Alazragi R., Alabdali A. Y. M., Aljazzar R., El Sadi S., Alostaz M., El Hindi M.. Antibacterial Activity of Metallic-Core Gold and Silver Nanoparticles against Some Animal Pathogens. Ann. Anim. Sci. 2023;23(2):473–479. doi: 10.2478/aoas-2023-0008. [DOI] [Google Scholar]
  558. Tripp R. A., Martin D. E.. Antiviral Agents and Therapeutics against Respiratory Viruses. Expert Opin. Invest. Drugs. 2024;33:1129–1133. doi: 10.1080/13543784.2024.2401911. [DOI] [PubMed] [Google Scholar]
  559. Abate C., Carnamucio F., Giuffrè O., Foti C.. Metal-Based Compounds in Antiviral Therapy. Biomolecules. 2022;12:933. doi: 10.3390/biom12070933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  560. Lysenko V., Lozovski V., Lokshyn M., Gomeniuk Y. V., Dorovskih A., Rusinchuk N., Pankivska Y., Povnitsa O., Zagorodnya S., Tertykh V., Bolbukh Y.. Nanoparticles as Antiviral Agents against Adenoviruses. Adv. Nat. Sci.: Nanosc. Nanotechnol. 2018;9(2):025021. doi: 10.1088/2043-6254/aac42a. [DOI] [Google Scholar]
  561. Chen L., Liang J.. An Overview of Functional Nanoparticles as Novel Emerging Antiviral Therapeutic Agents. Mater. Sci. Eng.: C. 2020;112:110924. doi: 10.1016/j.msec.2020.110924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  562. Sarkar P. K., Das Mukhopadhyay C.. Ayurvedic Metal Nanoparticles Could Be Novel Antiviral Agents against SARS-CoV-2. Int. Nano Lett. 2021;11(3):197–203. doi: 10.1007/s40089-020-00323-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  563. Sharmin S., Rahaman M. M., Sarkar C., Atolani O., Islam M. T., Adeyemi O. S.. Nanoparticles as Antimicrobial and Antiviral Agents: A Literature-Based Perspective Study. Heliyon. 2021;7(3):e06456. doi: 10.1016/j.heliyon.2021.e06456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  564. Ayipo Y. O., Bakare A. A., Badeggi U. M., Jimoh A. A., Lawal A., Mordi M. N.. Recent Advances on Therapeutic Potentials of Gold and Silver Nanobiomaterials for Human Viral Diseases. Curr. Res. Chem. Biol. 2022;2:100021. doi: 10.1016/j.crchbi.2022.100021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  565. Li X., Huang Y., Jin Q., Ji J.. Mixed-Charge Modification as a Robust Method to Realize the Antiviral Ability of Gold Nanoparticles in a High Protein Environment. Nanoscale. 2021;13(47):19857–19863. doi: 10.1039/D1NR06756G. [DOI] [PubMed] [Google Scholar]
  566. Chaika M., Zahorodnya S., Naumenko K., Pankivska Y., Povnitsa O., Mukha I., Vityuk N., Dorovskih A., Lokshyn M., Lysenko V., Lozovski V., Rusinchuk N.. Virus Deformation or Destruction: Size-Dependence of Antiviral and Virucidal Activities of Gold Nanoparticles. Adv. Nat. Sci.: Nanosci. Nanotechnol. 2022;13(3):035008. doi: 10.1088/2043-6262/ac879a. [DOI] [Google Scholar]
  567. Mehranfar A., Izadyar M.. Theoretical Design of Functionalized Gold Nanoparticles as Antiviral Agents against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) J. Phys. Chem. Lett. 2020;11(24):10284–10289. doi: 10.1021/acs.jpclett.0c02677. [DOI] [PubMed] [Google Scholar]
  568. Kesarkar R., Oza G., Pandey S., Dahake R., Mukherjee S., Chowdhary A., Sharon M.. Gold Nanoparticles: Effective as Both Entry Inhibitors and Virus Neutralizing Agents against HIV. J. Microbiol. Biotechnol. Res. 2012;2:276–283. [Google Scholar]
  569. Zacheo A., Hodek J., Witt D., Mangiatordi G. F., Ong Q. K., Kocabiyik O., Depalo N., Fanizza E., Laquintana V., Denora N., Migoni D., Barski P., Stellacci F., Weber J., Krol S.. Multi-Sulfonated Ligands on Gold Nanoparticles as Virucidal Antiviral for Dengue Virus. Sci. Rep. 2020;10(1):9052. doi: 10.1038/s41598-020-65892-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  570. Li C. M., Zheng L. L., Yang X. X., Wan X. Y., Wu W. B., Zhen S. J., Li Y. F., Luo L. F., Huang C. Z.. DNA-AuNP Networks on Cell Membranes as a Protective Barrier to Inhibit Viral Attachment, Entry and Budding. Biomaterials. 2016;77:216–226. doi: 10.1016/j.biomaterials.2015.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  571. Meléndez-Villanueva M. A., Morán-Santibañez K., Martínez-Sanmiguel J. J., Rangel-López R., Garza-Navarro M. A., Rodríguez-Padilla C., Zarate-Triviño D. G., Trejo-Ávila L. M.. Virucidal Activity of Gold Nanoparticles Synthesized by Green Chemistry Using Garlic Extract. Viruses. 2019;11(12):1111. doi: 10.3390/v11121111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  572. Ghaffari E., Rezatofighi S. E., Ardakani M. R., Rastegarzadeh S.. Delivery of Antisense Peptide Nucleic Acid by Gold Nanoparticles for the Inhibition of Virus Replication. Nanomedicine. 2019;14(14):1827–1840. doi: 10.2217/nnm-2018-0520. [DOI] [PubMed] [Google Scholar]
  573. Paradowska E., Studzińska M., Jabłońska A., Lozovski V., Rusinchuk N., Mukha I., Vitiuk N., Leśnikowski Z. J.. Antiviral Effect of Nonfunctionalized Gold Nanoparticles against Herpes Simplex Virus Type-1 (HSV-1) and Possible Contribution of Near-Field Interaction Mechanism. Molecules. 2021;26(19):5960. doi: 10.3390/molecules26195960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  574. El-Gaffary M., Bashandy M. M., Ahmed A. R., El-Borady O. M.. Self-Assembled Gold Nanoparticles for in-Vitro Inhibition of Bovine Viral Diarrhea Virus as Surrogate Model for HCV. Mater. Res. Express. 2019;6(7):075075. doi: 10.1088/2053-1591/ab18b6. [DOI] [Google Scholar]
  575. Lee M.-Y., Yang J.-A., Jung H. S., Beack S., Choi J. E., Hur W., Koo H., Kim K., Yoon S. K., Hahn S. K.. Hyaluronic Acid–Gold Nanoparticle/Interferon α Complex for Targeted Treatment of Hepatitis C Virus Infection. ACS Nano. 2012;6(11):9522–9531. doi: 10.1021/nn302538y. [DOI] [PubMed] [Google Scholar]
  576. Carja G., Grosu E. F., Petrarean C., Nichita N.. Self-Assemblies of Plasmonic Gold/Layered Double Hydroxides with Highly Efficient Antiviral Effect against the Hepatitis B Virus. Nano Res. 2015;8(11):3512–3523. doi: 10.1007/s12274-015-0851-6. [DOI] [Google Scholar]
  577. Krzyzowska M., Chodkowski M., Janicka M., Dmowska D., Tomaszewska E., Ranoszek-Soliwoda K., Bednarczyk K., Celichowski G., Grobelny J.. Lactoferrin-Functionalized Noble Metal Nanoparticles as New Antivirals for HSV-2 Infection. Microorganisms. 2022;10(1):110. doi: 10.3390/microorganisms10010110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  578. Bhebhe L. M., Jones L. M., Super E. H., Jones S. T.. Poly­(Styrene Sulfonate) Coated Gold Nanoparticles Display Virucidal Antiviral Activity. Polym. Sci. 2023:1–4. [Google Scholar]
  579. Montes-García V., Squillaci M. A., Diez-Castellnou M., Ong Q. K., Stellacci F., Samorì P.. Chemical Sensing with Au and Ag Nanoparticles. Chem. Soc. Rev. 2021;50(2):1269–1304. doi: 10.1039/D0CS01112F. [DOI] [PubMed] [Google Scholar]
  580. Jain P. K., Huang X., El-Sayed I. H., El-Sayed M. A.. Review of Some Interesting Surface Plasmon Resonance-Enhanced Properties of Noble Metal Nanoparticles and Their Applications to Biosystems. Plasmonics. 2007;2(3):107–118. doi: 10.1007/s11468-007-9031-1. [DOI] [Google Scholar]
  581. Seku, K. ; Hussaini, S. S. ; Reddy, M. R. ; Reddy, G. B. ; Kumar, K. K. . Fungal-Mediated Synthesis of Gold Nanoparticles and Their Biological Applications. In Fungal Cell Factories for Sustainable Nanomaterials Production and Agricultural Applications; Elsevier, 2022; pp 23–58 10.1016/B978-0-323-99922-9.00011-8. [DOI] [Google Scholar]
  582. Nejati K., Dadashpour M., Gharibi T., Mellatyar H., Akbarzadeh A.. Biomedical Applications of Functionalized Gold Nanoparticles: A Review. J. Cluster Sci. 2022;33:1–16. doi: 10.1007/s10876-020-01955-9. [DOI] [Google Scholar]
  583. Cho H. H., Jung D. H., Heo J. H., Lee C. Y., Jeong S. Y., Lee J. H.. Gold Nanoparticles as Exquisite Colorimetric Transducers for Water Pollutant Detection. ACS Appl. Mater. Interfaces. 2023;15(16):19785–19806. doi: 10.1021/acsami.3c00627. [DOI] [PubMed] [Google Scholar]
  584. Wang J., Cui W., Ren C. L., Ho E. A.. Ready-To-Use Microwave Sensor Modified by Antibody-AuNPs Nanoconjugate for Highly Sensitive and Selective Detection of the SARS-CoV-2 Virus. ACS Meas. Science Au. 2024;4(6):651–658. doi: 10.1021/acsmeasuresciau.4c00032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  585. Khan I. M., Niazi S., Yue L., Zhang Y., Pasha I., Iqbal Khan M. K., Akhtar W., Mohsin A., Chughati M. F. J., Wang Z.. Research Update of Emergent Gold Nanoclusters: A Reinforced Approach towards Evolution, Synthesis Mechanism and Application. Talanta. 2022;241:123228. doi: 10.1016/j.talanta.2022.123228. [DOI] [PubMed] [Google Scholar]
  586. Atta S., Vo-Dinh T.. Bimetallic Gold Nanostars Having High Aspect Ratio Spikes for Sensitive Surface-Enhanced Raman Scattering Sensing. ACS Appl. Nano Mater. 2022;5(9):12562–12570. doi: 10.1021/acsanm.2c02234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  587. Retout M., Brunetti E., Valkenier H., Bruylants G.. Limits of Thiol Chemistry Revealed by Quantitative Analysis of Mixed Layers of Thiolated-PEG Ligands Grafted onto Gold Nanoparticles. J. Colloid Interface Sci. 2019;557:807–815. doi: 10.1016/j.jcis.2019.09.047. [DOI] [PubMed] [Google Scholar]
  588. Moradi, S. ; Golbashy, M. ; Hosseini, M. . Sensing of Molecules by Electrocatalysis Based on Green Synthesis of Nanomaterials Top. Catal. 2025. 10.1007/s11244-025-02073-4. [DOI]
  589. Lee S. A., Kuhs C. T., Searles E. K., Everitt H. O., Landes C. F., Link S.. D-Band Hole Dynamics in Gold Nanoparticles Measured with Time-Resolved Emission Upconversion Microscopy. Nano Lett. 2023;23(8):3501–3506. doi: 10.1021/acs.nanolett.3c00622. [DOI] [PubMed] [Google Scholar]
  590. Chen Y., Cheng Y., Sun M.. Physical Mechanisms on Plasmon-Enhanced Organic Solar Cells. J. Phys. Chem. C. 2021;125(38):21301–21309. doi: 10.1021/acs.jpcc.1c07020. [DOI] [Google Scholar]
  591. Mandal P.. Application of Plasmonics in Solar Cell Efficiency Improvement: A Brief Review on Recent Progress. Plasmonics. 2022;17:1247–1267. doi: 10.1007/s11468-022-01616-9. [DOI] [Google Scholar]
  592. Ali A., El-Mellouhi F., Mitra A., Aïssa B.. Research Progress of Plasmonic Nanostructure-Enhanced Photovoltaic Solar Cells. Nanomaterials. 2022;12(5):788. doi: 10.3390/nano12050788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  593. Chi D., Lu S., Xu R., Liu K., Cao D., Wen L., Mi Y., Wang Z., Lei Y., Qu S., Wang Z.. Fully Understanding the Positive Roles of Plasmonic Nanoparticles in Ameliorating the Efficiency of Organic Solar Cells. Nanoscale. 2015;7(37):15251–15257. doi: 10.1039/C5NR04069H. [DOI] [PubMed] [Google Scholar]
  594. Islam O., Anik M. H. K., Mahmud S., Debnath J., Habib A., Islam S.. Optical, Thermal, and Electrical Analysis of Perovskite Solar Cell with Grated Cds and Embedded Plasmonic Au Nanoparticles. J. Phys. Energy. 2024;6(2):025007. doi: 10.1088/2515-7655/ad249b. [DOI] [Google Scholar]
  595. Aïssa B., Baloch A. A., Ali A., Mitra A.. Enhanced Multifaceted Model for Plasmon-Driven Schottky Solar Cells with Integrated Thermal Effects. Sci. Rep. 2025;15(1):3807. doi: 10.1038/s41598-024-82979-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  596. Rehman A., Rehman A. U., Aslam M., Muhammad N., Saeed M. A.. Optimization of Hollow Materials-Based Electron Transport Layer for Enhanced Performance of Perovskite Solar Cell. Plasmonics. 2024;19(3):1153–1164. doi: 10.1007/s11468-023-02057-8. [DOI] [Google Scholar]
  597. Tian J., Liu C., Forberich K., Barabash A., Xie Z., Qiu S., Byun J., Peng Z., Zhang K., Du T., Sathasivam S., Macdonald T. J., Dong L., Li C., Zhang J., Halik M., Le Corre V. M., Osvet A., Heumüller T., Li N., Zhou Y., Lüer L., Brabec C. J.. Overcoming Optical Losses in Thin Metal-Based Recombination Layers for Efficient n-i-p Perovskite-Organic Tandem Solar Cells. Nat. Commun. 2025;16(1):154. doi: 10.1038/s41467-024-55376-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  598. Kumar P., Huang S. H., Hsu C. Y., Chung S. Y., Cha H. C., Chuang C. M., Chen K. L., Huang Y. C.. Enhancing Power Conversion Efficiency of Organic Solar Cells with Magnetoplasmonic Fe3O4@Au@m-ABS Nanoparticles. Nanomaterials. 2024;14(14):1175. doi: 10.3390/nano14141175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  599. Gao H., Meng J., Sun J., Deng J.. Enhanced Performance of Polymer Solar Cells Based on P3HT:PCBM via Incorporating Au Nanoparticles Prepared by the Micellar Method. J. Mater. Sci.: Mater. Electron. 2020;31(13):10760–10767. doi: 10.1007/s10854-020-03626-x. [DOI] [Google Scholar]
  600. Merdeka M. C., Syarif N. F., Hidayat R., Wulandari P.. Incorporation of Gold Nanoparticle into ZnO Electron Transport Layer to Improve Performance of Hybrid Bulk Heterojunction Solar Cell. J. Phys.: Conf. Ser. 2024;2866:012016. doi: 10.1088/1742-6596/2866/1/012016. [DOI] [Google Scholar]
  601. Shin J., Song M., Hafeez H., Jeusraj P. J., Kim D. H., Lee J. C., Lee W. H., Choi D. K., Kim C. H., Bae T. S., Yu S. M., Kim K. H., Park H. G., Chung K. B., Song A., Kang Y. C., Park J., Kim C. S., Ryu S. Y.. Harvesting Near- and Far-Field Plasmonic Enhancements from Large Size Gold Nanoparticles for Improved Performance in Organic Bulk Heterojunction Solar Cells. Org. Electron. 2019;66:94–101. doi: 10.1016/j.orgel.2018.12.024. [DOI] [Google Scholar]
  602. Mohan M., Sekar R., Namboothiry M. A. G.. Understanding the Effects of Shape, Material and Location of Incorporation of Metal Nanoparticles on the Performance of Plasmonic Organic Solar Cells. RSC Adv. 2020;10(44):26126–26132. doi: 10.1039/D0RA04076B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  603. Kacus H., Baltakesmez A., Caldiran Z., Aydogan S., Yilmaz M., Sevim M.. Optical and Electrical Characterization of Organic Solar Cells Obtained Using Gold and Silver Metal Nanoparticles. Mater. Today: Proc. 2021;46:6986–6990. doi: 10.1016/j.matpr.2021.03.276. [DOI] [Google Scholar]
  604. Li Q., Wang F., Bai Y., Xu L., Yang Y., Yan L., Hu S., Zhang B., Dai S., Tan Z.. Decahedral-Shaped Au Nanoparticles as Plasmonic Centers for High Performance Polymer Solar Cells. Org. Electron. 2017;43:33–40. doi: 10.1016/j.orgel.2017.01.010. [DOI] [Google Scholar]
  605. Usmani B., Ranjan R., Prateek, Gupta S. K., Gupta R. K., Nalwa K. S., Garg A.. Inverted PTB7-Th:PC71BM Organic Solar Cells with 11.8% PCE via Incorporation of Gold Nanoparticles in ZnO Electron Transport Layer. Sol. Energy. 2021;214:220–230. doi: 10.1016/j.solener.2020.11.071. [DOI] [Google Scholar]
  606. Moongkarndi P., Rodpai E., Kanarat S.. Evaluation of an Immunochromatographic Assay for Rapid Detection of Salmonella Enterica Serovars Typhimurium and Enteritidis. J. Vet. Diagn. Invest. 2011;23(4):797–801. doi: 10.1177/1040638711408063. [DOI] [PubMed] [Google Scholar]
  607. Tan X., Ding S. Q., Hu Y. X., Li J. J., Zhou J. Y.. Development of an Immunosensor Assay for Detection of Haptoglobin in Mastitic Milk. Vet. Clin. Pathol. 2012;41(4):575–581. doi: 10.1111/j.1939-165X.2012.00468.x. [DOI] [PubMed] [Google Scholar]
  608. Kumar H., Kumar D., Valko M., Manickam S., Alomar S. Y., Cruz-Martins N., Kuča K.. Development of an Electrode Immunosensor Using Carbon Nanofibres-Gold Nanoparticles-Mercaptopropionic Acid-Polyethylenimine for Chicken Liver Containing Colistin. J. Agric. Food Res. 2024;15:100990. doi: 10.1016/j.jafr.2024.100990. [DOI] [Google Scholar]
  609. Farhan S. M., Al-Hadedee L. T.. The Effect of Gold Nano Rods on the Tissues of Mice and Their Use in Extending the Shelf Life of Chilled Minced Meat. IOP Conf. Ser.: Earth Environ. Sci. 2025;1449:012161. doi: 10.1088/1755-1315/1449/1/012161. [DOI] [Google Scholar]
  610. Loghmani A., Kamalpour M., Amrollahi-Sharifabadi M., Tapehbour M. B.. Targeted Nanoparticle-Based Therapy Using Betaine-Conjugated Gold Nanoparticles for Reducing Inflammation and Modulating Immune Response in Heatstroke-Induced Mice. J. Adv. Biomed. Sci. 2024;14:319–330. doi: 10.18502/jabs.v14i4.16692. [DOI] [Google Scholar]
  611. Peserico A., Canciello A., Prencipe G., Gramignoli R., Melai V., Scortichini G., Bellocci M., Capacchietti G., Turriani M., Di Pancrazio C., Berardinelli P., Russo V., Mattioli M., Barboni B.. Optimization of a Nanoparticle Uptake Protocol Applied to Amniotic-Derived Cells: Unlocking the Therapeutic Potential. J. Mater. Chem. B. 2024;12:8977–8992. doi: 10.1039/D4TB00607K. [DOI] [PubMed] [Google Scholar]
  612. Al-Maula B. H., Hussein B. J., Kadhim W. A., Sami S. M., Alamoush R. A., Abdulridh W. M., Ramalingam K., Zidan S., Al-Nasrawi S. J. H., Haider J., Peeran S. W.. The Effect of Gold Nanoparticles and Apricot Kernel Extract on the Osseointegration of Dental Implants - A Rabbit Model. Open Dent. J. 2024;18(1):e18742106311522. doi: 10.2174/0118742106311522240819071358. [DOI] [Google Scholar]
  613. Pournemati B., Tabesh H., Aghdam R. M., Rezayan A. H., Poorkhalil A., Tafti S. H. A., Heirani-Tabasi A., Eyni H., Malekmohamadi M., Boroumand S., Pinna A.. An Alginate/Gelatin Injectable Hydrogel Containing Au Nanoparticles for Transplantation of Embryonic Mouse Cardiomyocytes in Myocardial Repair. Macromol. Biosci. 2025;25:2400301. doi: 10.1002/mabi.202400301. [DOI] [PubMed] [Google Scholar]
  614. Geetha M., Singh A. K., Asokamani R., Gogia A. K.. Ti Based Biomaterials, the Ultimate Choice for Orthopaedic Implants - A Review. Prog. Mater. Sci. 2009;54:397–425. doi: 10.1016/j.pmatsci.2008.06.004. [DOI] [Google Scholar]
  615. Chen Q., Thouas G. A.. Metallic Implant Biomaterials. Mater. Sci. Eng. R: Rep. 2015;87:1–57. doi: 10.1016/j.mser.2014.10.001. [DOI] [Google Scholar]
  616. Basova T. V., Vikulova E. S., Dorovskikh S. I., Hassan A., Morozova N. B.. The Use of Noble Metal Coatings and Nanoparticles for the Modification of Medical Implant Materials. Mater. Des. 2021;204:109672. doi: 10.1016/j.matdes.2021.109672. [DOI] [Google Scholar]
  617. Jongrungsomran S., Pissuwan D., Yavirach A., Rungsiyakull C., Rungsiyakull P.. The Integration of Gold Nanoparticles into Dental Biomaterials as a Novel Approach for Clinical Advancement: A Narrative Review. J. Funct. Biomater. 2024;15:291. doi: 10.3390/jfb15100291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  618. Miyazawa N., Hakamada M., Mabuchi M.. Antimicrobial Mechanisms Due to Hyperpolarisation Induced by Nanoporous Au. Sci. Rep. 2018;8(1):3870. doi: 10.1038/s41598-018-22261-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  619. Solanki L. A., Sundari K. K. S., Rajeshkumar S.. In-Vitro Cytotoxicity Evaluation of Green Synthesized Gold Nanoparticles and Its Indigenous Mouthwash. J. Pure Appl. Microbiol. 2021;15(2):735–742. doi: 10.22207/JPAM.15.2.25. [DOI] [Google Scholar]
  620. Abuarqoub D., Mahmoud N., Alshaer W., Mohammad M., Ibrahim A. A., Al-Mrahleh M., Alnatour M., Alqudah D. A., Esawi E., Awidi A.. Biological Performance of Primary Dental Pulp Stem Cells Treated with Gold Nanoparticles. Biomedicines. 2023;11(9):2490. doi: 10.3390/biomedicines11092490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  621. Biz M. T., Cucco C., Cavalcanti B. N.. Incorporation of AuNP-PLL Nanocomplexes in DPSC: A New Tool for 3D Analysis in Pulp Regeneration. Clin Oral Invest. 2020;24(5):1761–1767. doi: 10.1007/s00784-019-03037-1. [DOI] [PubMed] [Google Scholar]
  622. Dharman S., Maragathavalli G., Shanmugam R., Shanmugasundaram K.. Curcumin Mediated Gold Nanoparticles and Analysis of Its Antioxidant, Anti-Inflammatory, Antimicrobial Activity Against Oral Pathogens. Pesqui. Bras. Odontopediatr. Clin. Integr. 2023;23:e220068. doi: 10.1590/pboci.2023.073. [DOI] [Google Scholar]
  623. Dalavi P. A., V A. J., Thomas S., Prabhu A., Anil S., Seong G. H., Venkatesan J.. Microwave-Assisted Biosynthesized Gold Nanoparticles Using Saussurea Obvallata: Biocompatibility and Antioxidant Activity Assessment. Bionanoscience. 2022;12(3):741–751. doi: 10.1007/s12668-022-00994-y. [DOI] [Google Scholar]
  624. Ivanovic V., Popovic D., Petrovic S., Rudolf R., Majerič P., Lazarevic M., Djordjevic I., Lazic V., Radunovic M.. Unraveling the Antibiofilm Activity of a New Nanogold Resin for Dentures and Epithesis. Pharmaceutics. 2022;14(7):1513. doi: 10.3390/pharmaceutics14071513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  625. Nam, K. Y. Characterization and Antifungal Activity of the Modified PMMA Denture Base Acrylic: Nanocomposites Impregnated with Gold, Platinum, and Silver Nanoparticles. In Nanobiomaterials in Dentistry: Applications of Nanobiomaterials; Elsevier, 2016; Vol. 11, pp 309–336 10.1016/B978-0-323-42867-5.00012-6. [DOI] [Google Scholar]
  626. Marin M., Nikolic M. V., Vidic J.. Rapid Point-of-Need Detection of Bacteria and Their Toxins in Food Using Gold Nanoparticles. Compr. Rev. Food Sci. Food Saf. 2021;20(6):5880–5900. doi: 10.1111/1541-4337.12839. [DOI] [PubMed] [Google Scholar]
  627. Hua Z., Yu T., Liu D., Xianyu Y.. Recent Advances in Gold Nanoparticles-Based Biosensors for Food Safety Detection. Biosens. Bioelectron. 2021;179:113076. doi: 10.1016/j.bios.2021.113076. [DOI] [PubMed] [Google Scholar]
  628. Viriyakitpattana N., Rattanabut C., Lertvachirapaiboon C., Pimalai D., Bamrungsap S.. Layer-by-Layer Biopolymer Assembly for the In Situ Fabrication of AuNP Plasmonic PaperA SERS Substrate for Food Adulteration Detection. ACS Omega. 2024;9(9):10099–10109. doi: 10.1021/acsomega.3c05966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  629. Ezhil Vilian A. T., Kang S.-M., Yeong Oh S., Woo Oh C., Umapathi R., Suk Huh Y., Han Y.-K.. A Simple Strategy for the Synthesis of Flower-like Textures of Au-ZnO Anchored Carbon Nanocomposite towards the High-performance Electrochemical Sensing of Sunset Yellow. Food Chem. 2020;323:126848. doi: 10.1016/j.foodchem.2020.126848. [DOI] [PubMed] [Google Scholar]
  630. Tariq, T. ; Saleem, I. M. ; Sahar, A. ; Raza, N. ; Sameen, A. ; Nayik, G. A. ; Ramniwas, S. . Nanogold Imprinted Starch Bionanocomposites for Food Packaging Applications. In Starch Based Nanomaterials for Food Packaging; Nayik, G. A. ; Hussain Dar, A. , Eds.; Academic Press, 2024; Chapter 7, pp 209–226 10.1016/B978-0-443-18967-8.00010-4. [DOI] [Google Scholar]
  631. Chandrababu V., Parameswaranpillai J., Gopi J. A., Pathak C., Midhun Dominic C. D., Feng N. L., Krishnasamy S., Muthukumar C., Hameed N., Ganguly S.. Progress in Food Packaging Applications of Biopolymer-Nanometal Composites  A Comprehensive Review. Biomater. Adv. 2024;162:213921. doi: 10.1016/j.bioadv.2024.213921. [DOI] [PubMed] [Google Scholar]
  632. Ahari H., Fakhrabadipour M., Paidari S., Goksen G., Xu B.. Role of AuNPs in Active Food Packaging Improvement: A Review. Molecules. 2022;27:8027. doi: 10.3390/molecules27228027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  633. Sadiq Z., Seyed Hamid S. T., Hasti H., Muna A.-K., Jahanshahi-Anbuhi S.. Gold Nanoparticles-Based Colorimetric Assays for Environmental Monitoring and Food Safety Evaluation. Crit Rev. Anal. Chem. 2024;54(7):2209–2244. doi: 10.1080/10408347.2022.2162331. [DOI] [PubMed] [Google Scholar]
  634. Liu G., Lu M., Huang X., Li T., Xu D.. Application of Gold-Nanoparticle Colorimetric Sensing to Rapid Food Safety Screening. Sensors. 2018;18:4166. doi: 10.3390/s18124166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  635. Ghosh S., Roy S., Naskar J., Kole R. K.. Plant-Mediated Synthesis of Mono- and Bimetallic (Au–Ag) Nanoparticles: Future Prospects for Food Quality and Safety. J. Nanomater. 2023;2023(1):2781667. doi: 10.1155/2023/2781667. [DOI] [Google Scholar]
  636. Sreelakshmi K. R., Mohan C. O., Remya S., Tejpal C. S., Ravishankar C. N.. Intrinsic Properties of Chitosan on the Characteristics of Gold Nanoparticles and Its Application as Smart Packaging Device. Food Sci. Technol. Int. 2024;30(2):169–181. doi: 10.1177/10820132221141944. [DOI] [PubMed] [Google Scholar]
  637. Alghamdi, A. M. Enhanced Optical, Mechanical, Thermal, and Antibacterial Properties in Polymer Bio-Nanocomposites Films via Loading Ag/Au Nanoparticles for Optical Devices and Food Packaging Polym. Compos. 2025; 10.1002/pc.29838. [DOI]
  638. Chowdhury S., Teoh Y. L., Ong K. M., Rafflisman Zaidi N. S., Mah S.-K.. Poly­(Vinyl) Alcohol Crosslinked Composite Packaging Film Containing Gold Nanoparticles on Shelf Life Extension of Banana. Food Packag. Shelf Life. 2020;24:100463. doi: 10.1016/j.fpsl.2020.100463. [DOI] [Google Scholar]
  639. Mehmood S., Janjua N. K., Tabassum S., Faizi S., Fenniri H.. Cost Effective Synthesis Approach for Green Food Packaging Coating by Gallic Acid Conjugated Gold Nanoparticles from Caesalpinia Pulcherrima Extract. Results Chem. 2022;4:100437. doi: 10.1016/j.rechem.2022.100437. [DOI] [Google Scholar]
  640. Yang C., Zhuang Z., Zou Y., Sun D., Zhang D., Liu X., Chen X.. A Novel Ratiometric Electrochemical Sensor Based on AuNPs Decorated MIL-101­(Fe) for Simultaneously Monitoring Typical EDCs in Milk. Sens Actuators, B. 2024;417:136224. doi: 10.1016/j.snb.2024.136224. [DOI] [Google Scholar]
  641. Sung J. H., Ji J. H., Park J. D., Song M. Y., Song K. S., Ryu H. R., Yoon J. U., Jeon K. S., Jeong J., Han B. S., Chung Y. H., Chang H. K., Lee J. H., Kim D. W., Kelman B. J., Yu I. J.. Subchronic Inhalation Toxicity of Gold Nanoparticles. Part. Fibre Toxicol. 2011;8:16. doi: 10.1186/1743-8977-8-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  642. Ramasamy, Ramasamy, Lee M., Lee J.. Development of Gold Nanoparticles Coated with Silica Containing the Antibiofilm Drug Cinnamaldehyde and Their Effects on Pathogenic Bacteria. Int. J. Nanomedicine. 2017;12:2813–2828. doi: 10.2147/IJN.S132784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  643. Elmetwalli A.. Impact of Antibiotic Interactions with Essential Oils on Bacterial Growth. J. Complem. Med. Res. 2022;13(3):114–116. doi: 10.5455/jcmr.2022.13.03.25. [DOI] [Google Scholar]
  644. Bagheri S., Maryam Y., Elmira S.-Q., Jamal R., Morteza A., Mahmoud H., Seyed Ali M., Kooshki H.. Using Gold Nanoparticles in Diagnosis and Treatment of Melanoma Cancer. Artif. Cells Nanomed. Biotechnol. 2018;46(sup1):462–471. doi: 10.1080/21691401.2018.1430585. [DOI] [PubMed] [Google Scholar]
  645. Chaudhry Q., Michael S., James B., Bryony R., Alistair B., Laurence C., Robert A., Watkins R.. Applications and Implications of Nanotechnologies for the Food Sector. Food Addit. Contam.,:Part A. 2008;25(3):241–258. doi: 10.1080/02652030701744538. [DOI] [PubMed] [Google Scholar]
  646. Jha P., Saraf A., Sohal J. K.. Antimicrobial Activity of Biologically Synthesized Gold Nanoparticles from Wild Mushroom Cantharellus Species. J. Sci. Res. 2021;65(03):78–83. doi: 10.37398/JSR.2021.650310. [DOI] [Google Scholar]
  647. Virgili A. H., Laranja D. C., Malheiros P. S., Pereira M. B., Costa T. M. H., de Menezes E. W.. Nanocomposite Film with Antimicrobial Activity Based on Gold Nanoparticles, Chitosan and Aminopropylsilane. Surf. Coat. Technol. 2021;415:127086. doi: 10.1016/j.surfcoat.2021.127086. [DOI] [Google Scholar]
  648. Glišić B. Đ., Djuran M. I.. Gold Complexes as Antimicrobial Agents: An Overview of Different Biological Activities in Relation to the Oxidation State of the Gold Ion and the Ligand Structure. Dalton Trans. 2014;43(16):5950–5969. doi: 10.1039/C4DT00022F. [DOI] [PubMed] [Google Scholar]
  649. MubarakAli D., Thajuddin N., Jeganathan K., Gunasekaran M.. Plant Extract Mediated Synthesis of Silver and Gold Nanoparticles and Its Antibacterial Activity against Clinically Isolated Pathogens. Colloids Surf., B. 2011;85(2):360–365. doi: 10.1016/j.colsurfb.2011.03.009. [DOI] [PubMed] [Google Scholar]
  650. Jia X., Yao Y., Yu G., Qu L., Li T., Li Z., Xu C.. Synthesis of Gold-Silver Nanoalloys under Microwave-Assisted Irradiation by Deposition of Silver on Gold Nanoclusters/Triple Helix Glucan and Antifungal Activity. Carbohydr. Polym. 2020;238:116169. doi: 10.1016/j.carbpol.2020.116169. [DOI] [PubMed] [Google Scholar]
  651. Hameed S., Wang Y., Zhao L., Xie L., Ying Y.. Shape-Dependent Significant Physical Mutilation and Antibacterial Mechanisms of Gold Nanoparticles against Foodborne Bacterial Pathogens (Escherichia Coli, Pseudomonas Aeruginosa and Staphylococcus Aureus) at Lower Concentrations. Mater. Sci. Eng.: C. 2020;108:110338. doi: 10.1016/j.msec.2019.110338. [DOI] [PubMed] [Google Scholar]
  652. Tavakolian S., Ahari H., Givianrad M. H., Hosseini H.. Improving the Barrier Properties of Food Packaging by Al2O3@TiO2&Al2O3@SiO2 Nanoparticles. Food Bioprocess Technol. 2021;14:1287–1300. doi: 10.1007/s11947-021-02635-w. [DOI] [Google Scholar]
  653. Pagno C. H., Costa T. M. H., de Menezes E. W., Benvenutti E. V., Hertz P. F., Matte C. R., Tosati J. V., Monteiro A. R., Rios A. O., Flôres S. H.. Development of Active Biofilms of Quinoa (Chenopodium Quinoa W.) Starch Containing Gold Nanoparticles and Evaluation of Antimicrobial Activity. Food Chem. 2015;173:755–762. doi: 10.1016/j.foodchem.2014.10.068. [DOI] [PubMed] [Google Scholar]
  654. Shabestarian H., Homayouni-Tabrizi M., Soltani M., Namvar F., Azizi S., Mohamad R., Shabestarian H.. Green Synthesis of Gold Nanoparticles Using Sumac Aqueous Extract and Their Antioxidant Activity. Mater. Res. 2017;20(1):264–270. doi: 10.1590/1980-5373-mr-2015-0694. [DOI] [Google Scholar]
  655. Maduraiveeran G., Ramaraj R.. Gold Nanoparticle-Based Sensing Platform of Hydrazine, Sulfite, and Nitrite for Food Safety and Environmental Monitoring. J. Anal. Sci. Technol. 2017;8(1):14. doi: 10.1186/s40543-017-0113-1. [DOI] [Google Scholar]
  656. G S., Jha P. K., V V., C R., M J., M S., Jha R., S S.. Cannonball Fruit (Couroupita Guianensis, Aubl.) Extract Mediated Synthesis of Gold Nanoparticles and Evaluation of Its Antioxidant Activity. J. Mol. Liq. 2016;215:229–236. doi: 10.1016/j.molliq.2015.12.043. [DOI] [Google Scholar]
  657. Ramamurthy C. H., Padma M., Samadanam I. D. M., Mareeswaran R., Suyavaran A., Kumar M. S., Premkumar K., Thirunavukkarasu C.. The Extra Cellular Synthesis of Gold and Silver Nanoparticles and Their Free Radical Scavenging and Antibacterial Properties. Colloids Surf., B. 2013;102:808–815. doi: 10.1016/j.colsurfb.2012.09.025. [DOI] [PubMed] [Google Scholar]
  658. Bindhu M. R., Umadevi M.. Silver and Gold Nanoparticles for Sensor and Antibacterial Applications. Spectrochim. Acta, Part A. 2014;128:37–45. doi: 10.1016/j.saa.2014.02.119. [DOI] [PubMed] [Google Scholar]
  659. Chen H., Zhou K., Zhao G.. Gold Nanoparticles: From Synthesis, Properties to Their Potential Application as Colorimetric Sensors in Food Safety Screening. Trends Food Sci. Technol. 2018;78:83–94. doi: 10.1016/j.tifs.2018.05.027. [DOI] [Google Scholar]
  660. Ferranti, P. Food Production and Ecosystem Protection. In Reference Module in Food Science; Elsevier, 2016. 10.1016/B978-0-08-100596-5.03444-2. [DOI] [Google Scholar]
  661. Chow C.-F.. Biogenic Amines- and Sulfides-Responsive Gold Nanoparticles for Real-Time Visual Detection of Raw Meat, Fish, Crustaceans, and Preserved Meat. Food Chem. 2020;311:125908. doi: 10.1016/j.foodchem.2019.125908. [DOI] [PubMed] [Google Scholar]
  662. Han C., Zhao A., Varughese E., Sahle-Demessie E.. Evaluating Weathering of Food Packaging Polyethylene-Nano-Clay Composites: Release of Nanoparticles and Their Impacts. NanoImpact. 2018;9:61–71. doi: 10.1016/j.impact.2017.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  663. Cushen M., Kerry J., Morris M., Cruz-Romero C.-R., Cummins E.. Silver Migration from Nanosilver and a Commercially Available Zeolite Filler Polyethylene Composites to Food Simulants. Food Addit. Contam.,:Part A. 2014;31(6):1132–1140. doi: 10.1080/19440049.2014.905874. [DOI] [PubMed] [Google Scholar]
  664. Zhang L., Mazouzi Y., Salmain M., Liedberg B., Boujday S.. Antibody-Gold Nanoparticle Bioconjugates for Biosensors: Synthesis, Characterization and Selected Applications. Biosens. Bioelectron. 2020;165:112370. doi: 10.1016/j.bios.2020.112370. [DOI] [PubMed] [Google Scholar]
  665. Awad M. A., Eisa N. E., Virk P., Hendi A. A., Ortashi K. M. O. O., Mahgoub A. S. A., Elobeid M. A., Eissa F. Z.. Green Synthesis of Gold Nanoparticles: Preparation, Characterization, Cytotoxicity, and Anti-Bacterial Activities. Mater. Lett. 2019;256:126608. doi: 10.1016/j.matlet.2019.126608. [DOI] [Google Scholar]
  666. Dasgupta N., Ranjan S., Mundekkad D., Ramalingam C., Shanker R., Kumar A.. Nanotechnology in Agro-Food: From Field to Plate. Food Res. Int. 2015;69:381–400. doi: 10.1016/j.foodres.2015.01.005. [DOI] [Google Scholar]
  667. Takeuchi M. T., Kojima M., Luetzow M.. State of the Art on the Initiatives and Activities Relevant to Risk Assessment and Risk Management of Nanotechnologies in the Food and Agriculture Sectors. Food Res. Int. 2014;64:976–981. doi: 10.1016/j.foodres.2014.03.022. [DOI] [PubMed] [Google Scholar]
  668. Marchiol L., Mattiello A., Pošćić F., Giordano C., Musetti R.. In Vivo Synthesis of Nanomaterials in Plants: Location of Silver Nanoparticles and Plant Metabolism. Nanoscale Res. Lett. 2014;9:101. doi: 10.1186/1556-276x-9-101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  669. Ribeiro C. A. S., Albuquerque L. J. C., de Castro C. E., Batista B. L., de Souza A. L. M., Albuquerque B. L., Zilse M. S., Bellettini I. C., Giacomelli F. C.. One-Pot Synthesis of Sugar-Decorated Gold Nanoparticles with Reduced Cytotoxicity and Enhanced Cellular Uptake. Colloids Surf., A. 2019;580:123690. doi: 10.1016/j.colsurfa.2019.123690. [DOI] [Google Scholar]
  670. Vijayaraghavan K., Ashokkumar T.. Plant-Mediated Biosynthesis of Metallic Nanoparticles: A Review of Literature, Factors Affecting Synthesis, Characterization Techniques and Applications. J. Environ. Chem. Eng. 2017;5(5):4866–4883. doi: 10.1016/j.jece.2017.09.026. [DOI] [Google Scholar]
  671. Prasad R., Bhattacharyya A., Nguyen Q. D.. Nanotechnology in Sustainable Agriculture: Recent Developments, Challenges, and Perspectives. Front. Microbiol. 2017;8:1014. doi: 10.3389/fmicb.2017.01014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  672. Sushma D., Richa S.. Use of Nanoparticles in Water Treatment: A Review. Int. Res. J. Environ. Sci. 2015;4:103–106. [Google Scholar]
  673. Omara A. E.-D., Elsakhawy T., Alshaal T., El-Ramady H., Kovács Z., Fári M.. Nanoparticles: A Novel Approach for Sustainable Agro-Productivity. Environ., Biodiversity Soil Secur. 2019;3(2019):30–40. doi: 10.21608/jenvbs.2019.7478.1050. [DOI] [Google Scholar]
  674. Fernández-Luqueño, F. ; Medina-Pérez, G. ; López-Valdez, F. ; Gutiérrez-Ramírez, R. ; Campos-Montiel, R. G. ; Vázquez-Núñez, E. ; Loera-Serna, S. ; Almaraz-Buendía, I. ; Del Razo-Rodríguez, O. E. ; Madariaga-Navarrete, A. . Use of Agronanobiotechnology in the Agro-Food Industry to Preserve Environmental Health and Improve the Welfare of Farmers. In Agricultural Nanobiotechnology: Modern Agriculture for a Sustainable Future; López-Valdez, F. ; Fernández-Luqueño, F. , Eds.; Springer International Publishing: Cham, 2018; pp 3–16 10.1007/978-3-319-96719-6_1. [DOI] [Google Scholar]
  675. Graily-Moradi, F. ; Mallak, A. M. ; Ghorbanpour, M. . Biogenic Synthesis of Gold Nanoparticles and Their Potential Application in Agriculture. In Biogenic Nano-Particles and their Use in Agro-Ecosystems; Ghorbanpour, M. ; Bhargava, P. ; Varma, A. ; Choudhary, D. K. , Eds.; Springer Singapore: Singapore, 2020; pp 187–204 10.1007/978-981-15-2985-6_11. [DOI] [Google Scholar]
  676. Xia M., Zhou J., Hu L., Li Y.. Highly Stable and Efficient Gold Nanoparticles Films Facilitated by Thiourea for Catalytic Reduction of Nitrophenol in Water. J. Environ. Chem. Eng. 2024;12(3):113004. doi: 10.1016/j.jece.2024.113004. [DOI] [Google Scholar]
  677. Francis S., Joseph S., Koshy E. P., Mathew B.. Green Synthesis and Characterization of Gold and Silver Nanoparticles Using Mussaenda Glabrata Leaf Extract and Their Environmental Applications to Dye Degradation. Environ. Sci. Pollut. Res. 2017;24(21):17347–17357. doi: 10.1007/s11356-017-9329-2. [DOI] [PubMed] [Google Scholar]
  678. Porwal, O. ; Singh, A. ; Singh, S. ; Fuloria, N. ; Patel, D. ; Chitranshi, N. ; Gupta, S. ; Varshney, P. . Patent Landscape of Nanopesticides, Nanoherbicides, and Nanofertilizers. In Nanopesticides, Nanoherbicides, and Nanofertilizers; Taylor and Francis, 2023; pp 137–162 10.1201/9781003364429-8. [DOI] [Google Scholar]
  679. Pathak, J. ; Singh, D. K. ; Singh, P. R. ; Kumari, N. ; Jaiswal, J. ; Gupta, A. ; Sinha, R. P. . Application of Nanoparticles in Agriculture: Nano-Based Fertilizers, Pesticides, Herbicides, and Nanobiosensors. In Molecular Impacts of Nanoparticles on Plants and Algae; Tombuloglu, H. ; Tombuloglu, G. ; Al-Suhaimi, E. ; Baykal, A. ; Hakeem, K. R. , Eds.; Academic Press, 2024; Chapter 14, pp 305–331 10.1016/B978-0-323-95721-2.00012-9. [DOI] [Google Scholar]
  680. Dhoundiyal, S. ; Kaur, A. ; Alam, M. A. ; Sharma, A. . The Future of Nanopesticides, Nanoherbicides, and Nanofertilizers. In Nanopesticides, Nanoherbicides, and Nanofertilizers; Taylor and Francis, 2023; pp 114–136 10.1201/9781003364429-7. [DOI] [Google Scholar]
  681. Kah M.. Nanopesticides and Nanofertilizers: Emerging Contaminants or Opportunities for Risk Mitigation. Front. Chem. 2015;3:64. doi: 10.3389/fchem.2015.00064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  682. Pereira, C. ; Pereira, A. M. ; Freire, C. ; Pinto, T. V. ; Costa, R. S. ; Teixeira, J. S. . Nanoengineered Textiles: From Advanced Functional Nanomaterials to Groundbreaking High-Performance Clothing. In Handbook of Functionalized Nanomaterials for Industrial Applications; Mustansar Hussain, C. , Ed.; Elsevier, 2020; Chapter 21, pp 611–714 10.1016/B978-0-12-816787-8.00021-1. [DOI] [Google Scholar]
  683. Mehravani B., Ribeiro A. I., Zille A.. Gold Nanoparticles Synthesis and Antimicrobial Effect on Fibrous Materials. Nanomaterials. 2021;11:1067. doi: 10.3390/nano11051067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  684. Eddy Jai Poinern G.. Gold Nanoparticle Treated Textile-Based Materials for Potential Use as Wearable Sensors. Int. J. Sci. 2016;2(05):82–89. doi: 10.18483/ijSci.1018. [DOI] [Google Scholar]
  685. Zheng Y., Xiao M., Jiang S., Ding F., Wang J.. Coating Fabrics with Gold Nanorods for Colouring, UV-Protection, and Antibacterial Functions. Nanoscale. 2013;5(2):788–795. doi: 10.1039/C2NR33064D. [DOI] [PubMed] [Google Scholar]
  686. Shanmugasundaram O. L., Ramkumar M.. Characterization and Study of Physical Properties and Antibacterial Activities of Human Hair Keratin–Silver Nanoparticles and Keratin–Gold Nanoparticles Coated Cotton Gauze Fabric. J. Ind. Text. 2018;47(5):798–814. doi: 10.1177/1528083716674904. [DOI] [Google Scholar]
  687. Lin X., Zou F., Chen X., Tang B.. Functional Modification of Nylon Fabrics Based on Noble Metal Nanoparticles. IOP Conf. Ser. Mater. Sci. Eng. 2017;231(1):012175. doi: 10.1088/1757-899X/231/1/012175. [DOI] [Google Scholar]
  688. Radić N., Obradović B. M., Kostić M., Dojčinović B., Hudcová M., Kuraica M. M., Černák M.. Deposition of Gold Nanoparticles on Polypropylene Nonwoven Pretreated by Dielectric Barrier Discharge and Diffuse Coplanar Surface Barrier Discharge. Plasma Chem. Plasma Process. 2013;33(1):201–218. doi: 10.1007/s11090-012-9414-8. [DOI] [Google Scholar]
  689. Ikegami M., Matsumoto T., Kobayashi Y., Jikihara Y., Nakayama T., Ohashi H., Honma T., Takei T., Haruta M.. Air Purification by Gold Catalysts Supported on PET Nonwoven Fabric. Appl. Catal., B. 2013;134–135:130–135. doi: 10.1016/j.apcatb.2012.12.018. [DOI] [Google Scholar]
  690. Cheng D., Bai X., He M., Wu J., Yang H., Ran J., Cai G., Wang X.. Polydopamine-Assisted Immobilization of Ag@AuNPs on Cotton Fabrics for Sensitive and Responsive SERS Detection. Cellulose. 2019;26(6):4191–4204. doi: 10.1007/s10570-019-02343-x. [DOI] [Google Scholar]
  691. Saldan I., Dobrovetska O., Sus L., Makota O., Pereviznyk O., Kuntyi O., Reshetnyak O.. Electrochemical Synthesis and Properties of Gold Nanomaterials. J. Solid State Electrochem. 2018;22:637–656. doi: 10.1007/s10008-017-3835-5. [DOI] [Google Scholar]
  692. Zhang K., Zhang J., Liu Y., Wang Z., Yan C., Song C., Gao C., Wu Y.. A NIR Laser Induced Self-Healing PDMS/Gold Nanoparticles Conductive Elastomer for Wearable Sensor. J. Colloid Interface Sci. 2021;599:360–369. doi: 10.1016/j.jcis.2021.04.117. [DOI] [PubMed] [Google Scholar]
  693. Ly T. N., Park S.. Wearable Strain Sensor for Human Motion Detection Based on Ligand-Exchanged Gold Nanoparticles. J. Ind. Eng. Chem. 2020;82:122–129. doi: 10.1016/j.jiec.2019.10.003. [DOI] [Google Scholar]
  694. Khorablou Z., Shahdost-Fard F., Razmi H.. Flexible and Highly Sensitive Methadone Sensor Based on Gold Nanoparticles/Polythiophene Modified Carbon Cloth Platform. Sens Actuators, B. 2021;344:130284. doi: 10.1016/j.snb.2021.130284. [DOI] [Google Scholar]
  695. Khan B., Riaz Z., u Shan Ahmad R., Khoo B. L.. Advancements in Wearable Sensors for Cardiovascular Disease Detection for Health Monitoring. Mater. Sci. Eng. R: Rep. 2024;159:100804. doi: 10.1016/j.mser.2024.100804. [DOI] [Google Scholar]
  696. Chen C., Zhou J., Li Z., Xu Y., Ran T., Gen J.. Wearable Electrochemical Biosensors for In Situ Pesticide Analysis from Crops. J. Electrochem. Soc. 2023;170(11):117512. doi: 10.1149/1945-7111/ad0b75. [DOI] [Google Scholar]
  697. Ketelsen B., Yesilmen M., Schlicke H., Noei H., Su C. H., Liao Y. C., Vossmeyer T.. Fabrication of Strain Gauges via Contact Printing: A Simple Route to Healthcare Sensors Based on Cross-Linked Gold Nanoparticles. ACS Appl. Mater. Interfaces. 2018;10(43):37374–37385. doi: 10.1021/acsami.8b12057. [DOI] [PubMed] [Google Scholar]
  698. Kaefer K., Krüger K., Schlapp F., Uzun H., Celiksoy S., Flietel B., Heimann A., Schroeder T., Kempski O., Sönnichsen C.. Implantable Sensors Based on Gold Nanoparticles for Continuous Long-Term Concentration Monitoring in the Body. Nano Lett. 2021;21(7):3325–3330. doi: 10.1021/acs.nanolett.1c00887. [DOI] [PubMed] [Google Scholar]
  699. Chen Y., Sun Y., Li Y., Wen Z., Peng X., He Y., Hou Y., Fan J., Zang G., Zhang Y.. A Wearable Non-Enzymatic Sensor for Continuous Monitoring of Glucose in Human Sweat. Talanta. 2024;278:126499. doi: 10.1016/j.talanta.2024.126499. [DOI] [PubMed] [Google Scholar]
  700. Peng B., Liu X., Yao Y., Ping J., Ying Y.. A Wearable and Capacitive Sensor for Leaf Moisture Status Monitoring. Biosens. Bioelectron. 2024;245:115804. doi: 10.1016/j.bios.2023.115804. [DOI] [PubMed] [Google Scholar]
  701. Wang S., Wu Y., Gu Y., Li T., Luo H., Li L. H., Bai Y., Li L., Liu L., Cao Y., Ding H., Zhang T.. Wearable Sweatband Sensor Platform Based on Gold Nanodendrite Array as Efficient Solid Contact of Ion-Selective Electrode. Anal. Chem. 2017;89(19):10224–10231. doi: 10.1021/acs.analchem.7b01560. [DOI] [PubMed] [Google Scholar]
  702. Dau H. G., Dang M. A., Luyen Van N., Nguy T. P., Ukita Y., Lien Truong T.. Gold Nanoparticles Enhanced Color Stability in Sweat Glucose Wearable Colorimetric Sensor Based on Automated Microfluidic Chip. Adv. Nat. Sci.: Nanosci. Nanotechnol. 2025;16(2):025002. doi: 10.1088/2043-6262/adb564. [DOI] [Google Scholar]
  703. Zhang X.. Gold Nanoparticles: Recent Advances in the Biomedical Applications. Cell Biochem. Biophys. 2015;72(3):771–775. doi: 10.1007/s12013-015-0529-4. [DOI] [PubMed] [Google Scholar]
  704. Sarfraz N., Khan I.. Plasmonic Gold Nanoparticles (AuNPs): Properties, Synthesis and Their Advanced Energy, Environmental and Biomedical Applications. Chem. - Asian J. 2021;16:720–742. doi: 10.1002/asia.202001202. [DOI] [PubMed] [Google Scholar]
  705. Oladipo I. C., Lateef A., Azeez M. A., Asafa T. B., Yekeen T. A., Ogunsona S. B., Irshad H. M., Abbas S. H.. Characterization and Biomedical Application of Phytosynthesized Gold Nanoparticles from Datura Stramonium Seed Extract. IOP Conf. Ser.: Mater. Sci. Eng. 2020;805:012021. doi: 10.1088/1757-899X/805/1/012021. [DOI] [Google Scholar]
  706. Clarance P., Luvankar B., Sales J., Khusro A., Agastian P., Tack J. C., Al Khulaifi M. M., AL-Shwaiman H. A., Elgorban A. M., Syed A., Kim H. J.. Green Synthesis and Characterization of Gold Nanoparticles Using Endophytic Fungi Fusarium Solani and Its In-Vitro Anticancer and Biomedical Applications. Saudi J. Biol. Sci. 2020;27(2):706–712. doi: 10.1016/j.sjbs.2019.12.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  707. Das R. K., Gogoi N., Babu P. J., Sharma P., Mahanta C., Bora U.. The Synthesis of Gold Nanoparticles Using Amaranthus Spinosus Leaf Extract and Study of Their Optical Properties. Adv. Mater. Phys. Chem. 2012;02(04):275–281. doi: 10.4236/ampc.2012.24040. [DOI] [Google Scholar]
  708. Hamza R. S. A., Habeeb M. A.. Synthesis and Tuning the Structural, Morphological and Dielectric Characteristics of PVA-CMC-SiO2–Cr2O3 Hybrid Nanostructures for Nanoelectronics Devices. Opt Quantum Electron. 2023;55(8):705. doi: 10.1007/s11082-023-04995-3. [DOI] [Google Scholar]
  709. Babajani N., Kaulen C., Homberger M., Mennicken M., Waser R., Simon U., Karthäuser S.. Directed Immobilization of Janus-AuNP in Heterometallic Nanogaps: A Key Step toward Integration of Functional Molecular Units in Nanoelectronics. J. Phys. Chem. C. 2014;118(46):27142–27149. doi: 10.1021/jp5085179. [DOI] [Google Scholar]
  710. Inberg A., Livshits P., Zalevsky Z., Shacham-Diamand Y.. Electroless Deposition of Silver Thin Films on Gold Nanoparticles Catalyst for Micro and Nanoelectronics Applications. Microelectron. Eng. 2012;98:570–573. doi: 10.1016/j.mee.2012.06.020. [DOI] [Google Scholar]
  711. Ruiz D. D., Cardos K. L., Soto G., Samano E. C.. Gold Nanostructures Based on DNA Origami Templates with Applications in Nanoelectronics and Plasmonics. MRS Adv. 2017;2:4017–4023. doi: 10.1557/adv.2018.177. [DOI] [Google Scholar]
  712. Thilagam R., Gnanamani A.. Preparation, Characterization and Stability Assessment of Keratin and Albumin Functionalized Gold Nanoparticles for Biomedical Applications. Appl. Nanosci. 2020;10(6):1879–1892. doi: 10.1007/s13204-020-01250-z. [DOI] [Google Scholar]
  713. Thomas S. R., Casini A.. N-Heterocyclic Carbenes as “Smart” Gold Nanoparticle Stabilizers: State-of-the Art and Perspectives for Biomedical Applications. J. Organomet. Chem. 2021;938:121743. doi: 10.1016/j.jorganchem.2021.121743. [DOI] [Google Scholar]
  714. Abu-Tahon M. A., Alshammari F. A., Shahhat I. M., Ghareib M., Abdallah W. E.. Eco-Friendly Synthesis, Characterization, and Biomedical Applications of Biosynthesized Bimetallic Silver-Gold Nanoparticles by Culture Supernatant of Aspergillus Niger. Appl. Biochem. Biotechnol. 2025;197:137–158. doi: 10.1007/s12010-024-05035-w. [DOI] [PubMed] [Google Scholar]
  715. Acay H.. Utilization of Morchella Esculenta-Mediated Green Synthesis Golden Nanoparticles in Biomedicine Applications. Prep. Biochem. Biotechnol. 2021;51(2):127–136. doi: 10.1080/10826068.2020.1799390. [DOI] [PubMed] [Google Scholar]
  716. Singh H., Du J., Singh P., Yi T. H.. Ecofriendly Synthesis of Silver and Gold Nanoparticles by Euphrasia Officinalis Leaf Extract and Its Biomedical Applications. Artif. Cells Nanomed. Biotechnol. 2018;46(6):1163–1170. doi: 10.1080/21691401.2017.1362417. [DOI] [PubMed] [Google Scholar]
  717. Arunachalam K. D., Annamalai S. K.. Chrysopogon Zizanioides Aqueous Extract Mediated Synthesis, Characterization of Crystalline Silver and Gold Nanoparticles for Biomedical Applications. Int. J. Nanomed. 2013;8:2375–2384. doi: 10.2147/IJN.S44076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  718. Manikandakrishnan M., Palanisamy S., Vinosha M., Kalanjiaraja B., Mohandoss S., Manikandan R., Tabarsa M., You S. G., Prabhu N. M.. Facile Green Route Synthesis of Gold Nanoparticles Using Caulerpa Racemosa for Biomedical Applications. J. Drug Delivery Sci. Technol. 2019;54:101345. doi: 10.1016/j.jddst.2019.101345. [DOI] [Google Scholar]
  719. Guerrero S., Herance J. R., Rojas S., Mena J. F., Gispert J. D., Acosta G. A., Albericio F., Kogan M. J.. Synthesis and in Vivo Evaluation of the Biodistribution of a 18F-Labeled Conjugate Gold-Nanoparticle-Peptide with Potential Biomedical Application. Bioconjugate Chem. 2012;23(3):399–408. doi: 10.1021/bc200362a. [DOI] [PubMed] [Google Scholar]
  720. Rajakumar G., Gomathi T., Rahuman A. A., Thiruvengadam M., Mydhili G., Kim S. H., Lee T. J., Chung I. M.. Biosynthesis and Biomedical Applications of Gold Nanoparticles Using Eclipta Prostrata Leaf Extract. Appl. Sci. 2016;6(8):722. doi: 10.3390/app6080222. [DOI] [Google Scholar]
  721. Sant D. G., Gujarathi T. R., Harne S. R., Ghosh S., Kitture R., Kale S., Chopade B. A., Pardesi K. R.. Adiantum Philippense L. Frond Assisted Rapid Green Synthesis of Gold and Silver Nanoparticles. J. Nanopart. 2013;2013:182320. doi: 10.1155/2013/182320. [DOI] [Google Scholar]
  722. Kiranda H. K., Mahmud R., Abubakar D., Zakaria Z. A.. Fabrication, Characterization and Cytotoxicity of Spherical-Shaped Conjugated Gold-Cockle Shell Derived Calcium Carbonate Nanoparticles for Biomedical Applications. Nanoscale Res. Lett. 2018;13(1):1. doi: 10.1186/s11671-017-2411-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  723. Lewis D. J., Pikramenou Z.. Lanthanide-Coated Gold Nanoparticles for Biomedical Applications. Coord. Chem. Rev. 2014;273-274:213–225. doi: 10.1016/j.ccr.2014.03.019. [DOI] [Google Scholar]
  724. Oishi M., Nagasaki Y.. Synthesis, Characterization, and Biomedical Applications of Core-Shell-Type Stimuli-Responsive Nanogels - Nanogel Composed of Poly­[2-(N,N-Diethylamino)­Ethyl Methacrylate] Core and PEG Tethered Chains. React. Funct. Polym. 2007;67:1311–1329. doi: 10.1016/j.reactfunctpolym.2007.07.009. [DOI] [Google Scholar]
  725. Niżnik Ł., Noga M., Kobylarz D., Frydrych A., Krośniak A., Kapka-Skrzypczak L., Jurowski K.. Gold Nanoparticles (AuNPs)Toxicity, Safety and Green Synthesis: A Critical Review. Int. J. Mol. Sci. 2024;24:4057. doi: 10.3390/ijms25074057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  726. Jiang M., Dai S., Wang B., Xie Z., Li J., Wang L., Li S., Tan Y., Tian B., Shu Q., Huang J.. Gold Nanoparticles Synthesized Using Melatonin Suppress Cadmium Uptake and Alleviate Its Toxicity in Rice. Environ. Sci. Nano. 2021;8(4):1042–1056. doi: 10.1039/D0EN01172J. [DOI] [Google Scholar]
  727. Ozcicek I., Aysit N., Cakici C., Aydeger A.. The Effects of Surface Functionality and Size of Gold Nanoparticles on Neuronal Toxicity, Apoptosis, ROS Production and Cellular/Suborgan Biodistribution. Mater. Sci. Eng., C. 2021;128:112308. doi: 10.1016/j.msec.2021.112308. [DOI] [PubMed] [Google Scholar]
  728. Nagtode V. S., Cardoza C., Yasin H. K. A., Mali S. N., Tambe S. M., Roy P., Singh K., Goel A., Amin P. D., Thorat B. R., Cruz J. N., Pratap A. P.. Green Surfactants (Biosurfactants): A Petroleum-Free Substitute for SustainabilityComparison, Applications, Market, and Future Prospects. ACS Omega. 2023;8(13):11674–11699. doi: 10.1021/acsomega.3c00591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  729. Sati A., Nandiwdekar O., Ratnaparkhi A., Doke R. B., Pinjari D. V., Mali S. N., Pratap A. P.. Bio-Based Alkyd–Polyesteramide–Polyurethane Coatings from Castor, Neem, and Karanja Oils with Inherent Antimicrobial Properties for Enhanced Hygiene. Coatings. 2025;15(4):370. doi: 10.3390/coatings15040370. [DOI] [Google Scholar]
  730. Abdulhaq N. A., Elnady D. A., Abo El-atta H. M., El-Morsi D. A., Gad El-Hak S. A.. Assessment of Reproductive Toxicity of Gold Nanoparticles and Its Reversibility in Male Albino Rats. Toxicol. Res. 2024;40(1):57–72. doi: 10.1007/s43188-023-00203-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  731. Chen Y. S., Hung Y. C., Liau I., Huang G. S.. Assessment of the in Vivo Toxicity of Gold Nanoparticles. Nanoscale Res. Lett. 2009;4(8):858–864. doi: 10.1007/s11671-009-9334-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  732. Sadauskas E., Jacobsen N. R., Danscher G., Stoltenberg M., Vogel U., Larsen A., Kreyling W., Wallin H.. Biodistribution of Gold Nanoparticles in Mouse Lung Following Intratracheal Instillation. Chem. Cent J. 2009;3(1):1–7. doi: 10.1186/1752-153X-3-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  733. Bahamonde J., Brenseke B., Chan M. Y., Kent R. D., Vikesland P. J., Prater M. R.. Gold Nanoparticle Toxicity in Mice and Rats: Species Differences. Toxicol. Pathol. 2018;46(4):431–443. doi: 10.1177/0192623318770608. [DOI] [PubMed] [Google Scholar]
  734. Lopez-Chaves C., Soto-Alvaredo J., Montes-Bayon M., Bettmer J., Llopis J., Sanchez-Gonzalez C.. Gold Nanoparticles: Distribution, Bioaccumulation and Toxicity. In Vitro and in Vivo Studies. Nanomedicine. 2018;14(1):1–12. doi: 10.1016/j.nano.2017.08.011. [DOI] [PubMed] [Google Scholar]
  735. Balasubramanian S. K., Jittiwat J., Manikandan J., Ong C. N., Yu L. E., Ong W. Y.. Biodistribution of Gold Nanoparticles and Gene Expression Changes in the Liver and Spleen after Intravenous Administration in Rats. Biomaterials. 2010;31(8):2034–2042. doi: 10.1016/j.biomaterials.2009.11.079. [DOI] [PubMed] [Google Scholar]
  736. Mukherjee S., Sau S., Madhuri D., Bollu V. S., Madhusudana K., Sreedhar B., Banerjee R., Patra C. R.. Green Synthesis and Characterization of Monodispersed Gold Nanoparticles: Toxicity Study, Delivery of Doxorubicin and Its Bio-Distribution in Mouse Model. J. Biomed. Nanotechnol. 2015;12:165–181. doi: 10.1166/jbn.2015.2141. [DOI] [PubMed] [Google Scholar]
  737. Daems N., Verlinden B., Van Hoecke K., Cardinaels T., Baatout S., Michiels C., Lucas S., Aerts A.. In Vivo Pharmacokinetics, Biodistribution and Toxicity of Antibody-Conjugated Gold Nanoparticles in Healthy Mice. J. Biomed. Nanotechnol. 2020;16(6):985–996. doi: 10.1166/jbn.2020.2928. [DOI] [PubMed] [Google Scholar]
  738. De Jong W. H., Hagens W. I., Krystek P., Burger M. C., Sips A. J. A. M., Geertsma R. E.. Particle Size-Dependent Organ Distribution of Gold Nanoparticles after Intravenous Administration. Biomaterials. 2008;29(12):1912–1919. doi: 10.1016/j.biomaterials.2007.12.037. [DOI] [PubMed] [Google Scholar]
  739. Bhatti R., Shakeel H., Malik K., Qasim M., Khan M. A., Ahmed N., Jabeen S.. Inorganic Nanoparticles: Toxic Effects, Mechanisms of Cytotoxicity and Phytochemical Interactions. Adv. Pharm. Bull. 2022;12:757–762. doi: 10.34172/apb.2022.077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  740. Al-Shaibani S. W., Alaarage W. K., Wahid M. H. A., Aljdaimi A. I.. Appraisal the Toxicity of CaO and Gold Nanoparticles in Wistar Rats: A Systematic Review. Open Access Res. J. Multidiscip. Stud. 2025;9(1):001–009. doi: 10.53022/oarjms.2025.9.1.0074. [DOI] [Google Scholar]
  741. Jakic K., Selc M., Razga F., Nemethova V., Mazancova P., Havel F., Sramek M., Zarska M., Proska J., Masanova V., Uhnakova I., Makovicky P., Novotova M., Vykoukal V., Babelova A.. Long-Term Accumulation, Biological Effects and Toxicity of BSA-Coated Gold Nanoparticles in the Mouse Liver, Spleen, and Kidneys. Int. J. Nanomed. 2024;19:4103–4120. doi: 10.2147/IJN.S443168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  742. Abd Elhameed H. A. H., Attia M. S., Mohamed A. A. A., Alexeree S. M. I., Behery E. I. E., Alagawany M., Farag M. R., Di Cerbo A., Azzam M. M., Mawed S. A.. The Role of Phthalocyanine-Gold Nanoconjugates (Pc-Au NCs) in Ameliorating the Hepatic and Renal Toxicity-Induced by Silver Nanoparticles (Ag NPs) in Male Rats. Biol. Trace Elem. Res. 2024;202:5637–5652. doi: 10.1007/s12011-024-04209-1. [DOI] [PubMed] [Google Scholar]
  743. Isoda K., Tanaka A., Fuzimori C., Echigoya M., Taira Y., Taira I., Shimizu Y., Akimoto Y., Kawakami H., Ishida I.. Toxicity of Gold Nanoparticles in Mice Due to Nanoparticle/Drug Interaction Induces Acute Kidney Damage. Nanoscale Res. Lett. 2020;15(1):141. doi: 10.1186/s11671-020-03371-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  744. Zhang X. D., Wu H. Y., Wu D., Wang Y. Y., Chang J. H., Zhai Z., Bin, Meng A. M., Liu P. X., Zhang L. A., Fan F. Y.. Toxicologic Effects of Gold Nanoparticles in Vivo by Different Administration Routes. Int. J. Nanomed. 2010;5(1):771–781. doi: 10.2147/IJN.S8428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  745. Alansari W. S.. Sulforaphane (4-Methylsulfnylbutyl Isothiocyanate) Mitigates Gold Nanoparticle Induced Brain Toxicity in Male Albino Rats. J. King Saud Univ Sci. 2024;36(7):103257. doi: 10.1016/j.jksus.2024.103257. [DOI] [Google Scholar]
  746. Raji V., Kumar J., Rejiya C. S., Vibin M., John A., Abraham A.. Synthesis, Characterisation and Biocompatibility of Surface-Functionalised Gold Nanoparticles. J. Exp. Nanosci. 2012;7(2):174–188. doi: 10.1080/17458080.2010.514952. [DOI] [Google Scholar]
  747. Lasagna-Reeves C., Gonzalez-Romero D., Barria M. A., Olmedo I., Clos A., Ramanujam V. M. S., Urayama A., Vergara L., Kogan M. J., Soto C.. Bioaccumulation and Toxicity of Gold Nanoparticles after Repeated Administration in Mice. Biochem. Biophys. Res. Commun. 2010;393(4):649–655. doi: 10.1016/j.bbrc.2010.02.046. [DOI] [PubMed] [Google Scholar]
  748. Bar-Ilan O., Albrecht R. M., Fako V. E., Furgeson D. Y.. Toxicity Assessments of Multisized Gold and Silver Nanoparticles in Zebrafish Embryos. Small. 2009;5(16):1897–1910. doi: 10.1002/smll.200801716. [DOI] [PubMed] [Google Scholar]
  749. Ramachandran R., Krishnaraj C., Kumar V. K. A., Harper S. L., Kalaichelvan T. P., Yun S.. Il. In Vivo Toxicity Evaluation of Biologically Synthesized Silver Nanoparticles and Gold Nanoparticles on Adult Zebrafish: A Comparative Study. 3 Biotech. 2018;8(10):441. doi: 10.1007/s13205-018-1457-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  750. Kim K. T., Zaikova T., Hutchison J. E., Tanguay R. L.. Gold Nanoparticles Disrupt Zebrafish Eye Development and Pigmentation. Toxicol. Sci. 2013;133(2):275–288. doi: 10.1093/toxsci/kft081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  751. Botteon C. E. A., Pereira A., do, de Castro E. S., Justino L. P., Fraceto I. A., Bastos L. F., Marcato J. K.. P. D. Toxicity Assessment of Biogenic Gold Nanoparticles on Crop Seeds and Zebrafish Embryos: Implications for Agricultural and Aquatic Ecosystems. ACS Omega. 2025;10(1):1032–1046. doi: 10.1021/acsomega.4c08287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  752. Ibrahim B., Akere T. H., Dhumal P., Valsami-Jones E., Chakraborty S.. Designing Safer Nanohybrids: Stability and Ecotoxicological Assessment of Graphene Oxide-Gold Nanoparticle Hybrids in Embryonic Zebrafish. Environ. Sci.: Nano. 2025;12:1965–1978. doi: 10.1039/D4EN01173B. [DOI] [Google Scholar]
  753. Machado S., González-Ballesteros N., Gonçalves A., Magalhães L., de Passos M. S. P., Rodríguez-Argüelles M. C., Gomes A. C.. Toxicity in Vitro and in Zebrafish Embryonic Development of Gold Nanoparticles Biosynthesized Using Cystoseira Macroalgae Extracts. Int. J. Nanomed. 2021;16:5017–5036. doi: 10.2147/IJN.S300674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  754. Ramachandran R., Krishnaraj C., Sivakumar A. S., Prasannakumar P., Abhay Kumar V. K., Shim K. S., Song C. G., Yun S.. Il. Anticancer Activity of Biologically Synthesized Silver and Gold Nanoparticles on Mouse Myoblast Cancer Cells and Their Toxicity against Embryonic Zebrafish. Mater. Sci. Eng., C. 2017;73:674–683. doi: 10.1016/j.msec.2016.12.110. [DOI] [PubMed] [Google Scholar]
  755. Windell D. L., Mourabit S., Moger J., Owen S. F., Winter M. J., Tyler C. R.. The Influence of Size and Surface Chemistry on the Bioavailability, Tissue Distribution and Toxicity of Gold Nanoparticles in Zebrafish (Danio Rerio) Ecotoxicol. Environ. Saf. 2023;260:115019. doi: 10.1016/j.ecoenv.2023.115019. [DOI] [PubMed] [Google Scholar]
  756. Browning L. M., Lee K. J., Huang T., Nallathamby P. D., Lowman J. E., Nancy Xu X. H.. Random Walk of Single Gold Nanoparticles in Zebrafish Embryos Leading to Stochastic Toxic Effects on Embryonic Developments. Nanoscale. 2009;1(1):138–152. doi: 10.1039/b9nr00053d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  757. Javed I., Peng G., Xing Y., Yu T., Zhao M., Kakinen A., Faridi A., Parish C. L., Ding F., Davis T. P., Ke P. C., Lin S.. Inhibition of Amyloid Beta Toxicity in Zebrafish with a Chaperone-Gold Nanoparticle Dual Strategy. Nat. Commun. 2019;10(1):3780. doi: 10.1038/s41467-019-11762-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  758. Browning L. M., Huang T., Xu X. H. N.. Real-Time in Vivo Imaging of Sizedependent Transport and Toxicity of Gold Nanoparticles in Zebrafish Embryos Using Single Nanoparticle Plasmonic Spectroscopy. Interface Focus. 2013;3(3):20120098. doi: 10.1098/rsfs.2012.0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  759. Suman T. Y., Jia H. J., Yin S., Wei X. Y., Hu H., Bu L. K., Yang G., Pei D. S.. Biofabricated Gold Nanoparticles from Ammannia Baccifera as Potential Antimicrobial, Mosquito Larvicidal Activity, and Alter Immune Response in Zebrafish Embryo. Biomass Convers. Biorefin. 2024;14(18):22967–22976. doi: 10.1007/s13399-023-04360-0. [DOI] [Google Scholar]
  760. Ganeshkumar M., Sastry T. P., Kumar M. S., Dinesh M. G., Kannappan S., Suguna L.. Sun Light Mediated Synthesis of Gold Nanoparticles as Carrier for 6-Mercaptopurine: Preparation, Characterization and Toxicity Studies in Zebrafish Embryo Model. Mater. Res. Bull. 2012;47(9):2113–2119. doi: 10.1016/j.materresbull.2012.06.015. [DOI] [Google Scholar]
  761. Sowmiya P., Dhas T. S., Sai D. N., Jyotsna, Anandakumar N., Kumar V. G., Ravi M., Nalini S., Borgio J. F., AbdulAzeez S., Karthick V.. Genotoxicity Evaluation of Pectin-Mediated Gold Nanoparticles on Zebrafish Embryos (Danio Rerio) Appl. Biochem. Microbiol. 2022;58(2):186–194. doi: 10.1134/S0003683822020156. [DOI] [Google Scholar]
  762. Vetten M. A., Tlotleng N., Tanner Rascher D., Skepu A., Keter F. K., Boodhia K., Koekemoer L. A., Andraos C., Tshikhudo R., Gulumian M.. Label-Free in Vitro Toxicity and Uptake Assessment of Citrate Stabilised Gold Nanoparticles in Three Cell Lines. Part. Fibre Toxicol. 2013;10(1):50. doi: 10.1186/1743-8977-10-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  763. Uboldi C., Bonacchi D., Lorenzi G., Iris M. I., Pohl C., Baldi G., Unger R. E., James C. J.. Gold Nanoparticles Induce Cytotoxicity in the Alveolar Type-II Cell Lines A549 and NCIH441. Part. Fibre Toxicol. 2009;6:18. doi: 10.1186/1743-8977-6-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  764. Coulter J. A., Jain S., Butterworth K. T., Taggart L. E., Dickson G. R., McMahon S. J., Hyland W. B., Muir M. F., Trainor C., Hounsell A. R., O’Sullivan J. M., Schettino G., Currell F. J., Hirst D. G., Prise K. M.. Cell Type-Dependent Uptake, Localization, and Cytotoxicity of 1.9 Nm Gold Nanoparticles. Int. J. Nanomed. 2012;7:2673–2685. doi: 10.2147/IJN.S31751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  765. Chueh P. J., Liang R. Y., Lee Y. H., Zeng Z. M., Chuang S. M.. Differential Cytotoxic Effects of Gold Nanoparticles in Different Mammalian Cell Lines. J. Hazard. Mater. 2014;264:303–312. doi: 10.1016/j.jhazmat.2013.11.031. [DOI] [PubMed] [Google Scholar]
  766. Schneider T., Westermann M., Glei M.. In Vitro Uptake and Toxicity Studies of Metal Nanoparticles and Metal Oxide Nanoparticles in Human HT29 Cells. Arch. Toxicol. 2017;91(11):3517–3527. doi: 10.1007/s00204-017-1976-z. [DOI] [PubMed] [Google Scholar]
  767. Al-Radadi N. S.. Green Biosynthesis of Flaxseed Gold Nanoparticles (Au-NPs) as Potent Anti-Cancer Agent Against Breast Cancer Cells. J. Saudi Chem. Soc. 2021;25(6):101243. doi: 10.1016/j.jscs.2021.101243. [DOI] [Google Scholar]
  768. Das S., Debnath N., Mitra S., Datta A., Goswami A.. Comparative Analysis of Stability and Toxicity Profile of Three Differently Capped Gold Nanoparticles for Biomedical Usage. BioMetals. 2012;25(5):1009–1022. doi: 10.1007/s10534-012-9567-1. [DOI] [PubMed] [Google Scholar]
  769. Vedantam P., Huang G., Tzeng T. R. J.. Size-Dependent Cellular Toxicity and Uptake of Commercial Colloidal Gold Nanoparticles in DU-145 Cells. Cancer Nanotechnol. 2013;4(1–3):13–20. doi: 10.1007/s12645-013-0033-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  770. Iqbal Y., Amin F., Aziz M. H., Alhadlaq H. A., Alaizeri Z. A. M.. Biomimetic Sodium Alginate Functionalized Gold Nanoparticles Loaded with Trans-Resveratrol for in-Vitro Cancer Treatment and Intercellular ROS Studies against MCF-7 Cell Lines. Colloids Surf., A. 2025;712:136448. doi: 10.1016/j.colsurfa.2025.136448. [DOI] [Google Scholar]
  771. Choi K., Joo H.. Assessment of Gold Nanoparticles-Inhibited Cytochrome P450 3A4 Activity and Molecular Mechanisms Underlying Its Cellular Toxicity in Human Hepatocellular Carcinoma Cell Line C3A. Nanoscale Res. Lett. 2018;13:279. doi: 10.1186/s11671-018-2684-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  772. Das A. K., Borah M., Kalita J. J., Bora U.. Cytotoxic Potential of Curcuma Caesia Rhizome Extract and Derived Gold Nanoparticles in Targeting Breast Cancer Cell Lines. Sci. Rep. 2024;14(1):17223. doi: 10.1038/s41598-024-66175-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  773. Sereemaspun, A. ; Pongsuchart, M. ; Danladkaew, C. ; Khomvarn, T. In Effect of Glutathione-Stabilized Gold Nanoparticles in 3T3 Fibroblast Cell, 2012 International Conference on Clean and Green Energy; IACSIT, 2012. [Google Scholar]
  774. Mousavi-Kouhi S. M., Beyk-Khormizi A., Mohammadzadeh V., Ashna M., Es-haghi A., Mashreghi M., Hashemzadeh V., Mozafarri H., Nadaf M., Taghavizadeh Yazdi M. E.. Biological Synthesis and Characterization of Gold Nanoparticles Using Verbascum Speciosum Schrad. and Cytotoxicity Properties toward HepG2 Cancer Cell Line. Res. Chem. Intermed. 2022;48(1):167–178. doi: 10.1007/s11164-021-04600-w. [DOI] [Google Scholar]
  775. ul G Mir, T. ; Katoch, V. ; Angurana, R. ; Wani, A. K. ; Shukla, S. ; El Messaoudi, N. ; Sher, F. ; Mulla, S. I. ; Américo-Pinheiro, J. H. P. . Environmental and Toxicological Concerns Associated with Nanomaterials Used in the Industries. In Nanomaterials for Bioreactors and Bioprocessing Applications; Elsevier, 2023; pp 141–193 10.1016/B978-0-323-91782-7.00010-2. [DOI] [Google Scholar]
  776. Balashanmugam P., Durai P., Balakumaran M. D., Kalaichelvan P. T.. Phytosynthesized Gold Nanoparticles from C. Roxburghii DC. Leaf and Their Toxic Effects on Normal and Cancer Cell Lines. J. Photochem. Photobiol. B. 2016;165:163–173. doi: 10.1016/j.jphotobiol.2016.10.013. [DOI] [PubMed] [Google Scholar]
  777. Abdoli M., Arkan E., Shekarbeygi Z., Khaledian S.. Green Synthesis of Gold Nanoparticles Using Centaurea Behen Leaf Aqueous Extract and Investigating Their Antioxidant and Cytotoxic Effects on Acute Leukemia Cancer Cell Line (THP-1) Inorg. Chem. Commun. 2021;129:108649. doi: 10.1016/j.inoche.2021.108649. [DOI] [Google Scholar]
  778. Mahmoud N. N., Salman T. M., Al-Dabash S., Abdullah M., Abu-Dahab R.. The Impact of Gold Nanoparticles Conjugated with Albumin on Prostate and Breast Cancer Cell Lines: Insights into Cytotoxicity, Cellular Uptake, Migration, and Adhesion Potential. J. Nanopart. Res. 2024;26(5):101. doi: 10.1007/s11051-024-05990-9. [DOI] [Google Scholar]
  779. Reipa V., Hackley V. A., Tona A., Heo M. B., Lee Y. R., Lee T. G., Johnston-Peck A., Cho T. J.. Well-Characterized Polyethyleneimine-/Carboxylated-Polyethylene-Glycol-Functionalized Gold Nanoparticles as Prospective Nanoscale Control Materials for In Vitro Cell Viability Assays: Particle Characterization and Toxicity Tests in Eight Mammalian Cell Lines. Nanomaterials. 2025;15(2):79. doi: 10.3390/nano15020079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  780. Trejo-Teniente I., Jaramillo-Loranca B. E., Vargas-Hernández G., Villanueva-Ibáñez M., Tovar-Jiménez X., Olvera-Venegas P. N., Tapia-Ramírez J.. Synthesis and Toxicity Assessment of Coffea Arabica Extract-Derived Gold Nanoparticles Loaded with Doxorubicin in Lung Cancer Cell Cultures. Front. Bioeng. Biotechnol. 2024;12:1378601. doi: 10.3389/fbioe.2024.1378601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  781. Nandhini J. T., Ezhilarasan D., Rajeshkumar S.. An Ecofriendly Synthesized Gold Nanoparticles Induces Cytotoxicity via Apoptosis in HepG2 Cells. Environ. Toxicol. 2021;36(1):24–32. doi: 10.1002/tox.23007. [DOI] [PubMed] [Google Scholar]
  782. Enea M., Pereira E., Costa J., Soares M. E., da Silva D. D., Bastos M., de Lourdes Bastos M., Carmo L.. H. F. Cellular Uptake and Toxicity of Gold Nanoparticles on Two Distinct Hepatic Cell Models. Toxicol. in Vitro. 2021;70:105046. doi: 10.1016/j.tiv.2020.105046. [DOI] [PubMed] [Google Scholar]
  783. Pan Y., Leifert A., Ruau D., Neuss S., Bornemann J., Schmid G., Brandau W., Simon U., Jahnen-Dechent W.. Gold Nanoparticles of Diameter 1.4 Nm Trigger Necrosis by Oxidative Stress and Mitochondrial Damage. Small. 2009;5(18):2067–2076. doi: 10.1002/smll.200900466. [DOI] [PubMed] [Google Scholar]
  784. Coradeghini R., Gioria S., García C. P., Nativo P., Franchini F., Gilliland D., Ponti J., Rossi F.. Size-Dependent Toxicity and Cell Interaction Mechanisms of Gold Nanoparticles on Mouse Fibroblasts. Toxicol. Lett. 2013;217(3):205–216. doi: 10.1016/j.toxlet.2012.11.022. [DOI] [PubMed] [Google Scholar]
  785. Boyoglu C., He Q., Willing G., Boyoglu-Barnum S., Dennis V. A., Pillai S., Singh S. R.. Microscopic Studies of Various Sizes of Gold Nanoparticles and Their Cellular Localizations. ISRN Nanotechnol. 2013;2013:123838. doi: 10.1155/2013/123838. [DOI] [Google Scholar]
  786. Shang L., Nienhaus K., Nienhaus G. U.. Engineered Nanoparticles Interacting with Cells: Size Matters. J. Nanobiotechnol. 2014;12:5. doi: 10.1186/1477-3155-12-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  787. Bhamidipati M., Fabris L.. Multiparametric Assessment of Gold Nanoparticle Cytotoxicity in Cancerous and Healthy Cells: The Role of Size, Shape, and Surface Chemistry. Bioconjugate Chem. 2017;28(2):449–460. doi: 10.1021/acs.bioconjchem.6b00605. [DOI] [PubMed] [Google Scholar]
  788. Alkilany A. M., Nagaria P. K., Hexel C. R., Shaw T. J., Murphy C. J., Wyatt M. D.. Cellular Uptake and Cytotoxicity of Gold Nanorods: Molecular Origin of Cytotoxicity and Surface Effects. Small. 2009;5(6):701–708. doi: 10.1002/smll.200801546. [DOI] [PubMed] [Google Scholar]
  789. Nelson C. E., Kintzing J. R., Hanna A., Shannon J. M., Gupta M. K., Duvall C. L.. Balancing Cationic and Hydrophobic Content of PEGylated SiRNA Polyplexes Enhances Endosome Escape, Stability, Blood Circulation Time, and Bioactivity in Vivo. ACS Nano. 2013;7(10):8870–8880. doi: 10.1021/nn403325f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  790. Jin E., Zhang B., Sun X., Zhou Z., Ma X., Sun Q., Tang J., Shen Y., Van Kirk E., Murdoch W. J., Radosz M.. Acid-Active Cell-Penetrating Peptides for in Vivo Tumor-Targeted Drug Delivery. J. Am. Chem. Soc. 2013;135(2):933–940. doi: 10.1021/ja311180x. [DOI] [PubMed] [Google Scholar]
  791. De La Fuente J. M., Berry C. C.. Tat Peptide as an Efficient Molecule To Translocate Gold Nanoparticles into the Cell Nucleus. Bioconjugate Chem. 2005;16:1176–1180. doi: 10.1021/bc050033. [DOI] [PubMed] [Google Scholar]
  792. Elsayed K. A., Imam H., Ahmed M. A., Ramadan R.. Effect of Focusing Conditions and Laser Parameters on the Fabrication of Gold Nanoparticles via Laser Ablation in Liquid. Opt. Laser Technol. 2013;45(1):495–502. doi: 10.1016/j.optlastec.2012.06.004. [DOI] [Google Scholar]
  793. Correard F., Maximova K., Estève M. A., Villard C., Roy M., Al-Kattan A., Sentis M., Gingras M., Kabashin A. V., Braguer D.. Gold Nanoparticles Prepared by Laser Ablation in Aqueous Biocompatible Solutions: Assessment of Safety and Biological Identity for Nanomedicine Applications. Int. J. Nanomed. 2014;9(1):5415–5430. doi: 10.2147/IJN.S65817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  794. Boyles M. S. P., Kristl T., Andosch A., Zimmermann M., Tran N., Casals E., Himly M., Puntes V., Huber C. G., Lütz-Meindl U., Duschl A.. Chitosan Functionalisation of Gold Nanoparticles Encourages Particle Uptake and Induces Cytotoxicity and Pro-Inflammatory Conditions in Phagocytic Cells, as Well as Enhancing Particle Interactions with Serum Components. J. Nanobiotechnol. 2015;13(1):84. doi: 10.1186/s12951-015-0146-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  795. Moore T. L., Rodriguez-Lorenzo L., Hirsch V., Balog S., Urban D., Jud C., Rothen-Rutishauser B., Lattuada M., Petri-Fink A.. Nanoparticle Colloidal Stability in Cell Culture Media and Impact on Cellular Interactions. Chem. Soc. Rev. 2015;44(17):6287–6305. doi: 10.1039/C4CS00487F. [DOI] [PubMed] [Google Scholar]
  796. Yang J. A., Lohse S. E., Murphy C. J.. Tuning Cellular Response to Nanoparticles via Surface Chemistry and Aggregation. Small. 2014;10(8):1642–1651. doi: 10.1002/smll.201302835. [DOI] [PubMed] [Google Scholar]
  797. Yu M., Zheng J.. Clearance Pathways and Tumor Targeting of Imaging Nanoparticles. ACS Nano. 2015;9:6655–6674. doi: 10.1021/acsnano.5b01320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  798. García-Álvarez R., Hadjidemetriou M., Sánchez-Iglesias A., Liz-Marzán L. M., Kostarelos K.. In Vivo Formation of Protein Corona on Gold Nanoparticles. the Effect of Their Size and Shape. Nanoscale. 2018;10(3):1256–1264. doi: 10.1039/C7NR08322J. [DOI] [PubMed] [Google Scholar]
  799. Gunduz N., Ceylan H., Guler M. O., Tekinay A. B.. Intracellular Accumulation of Gold Nanoparticles Leads to Inhibition of Macropinocytosis to Reduce the Endoplasmic Reticulum Stress. Sci. Rep. 2017;7:40493. doi: 10.1038/srep40493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  800. Stewart M., Mulenos M. R., Steele L. R., Sayes C. M.. Differences among Unique Nanoparticle Protein Corona Constructs: A Case Study Using Data Analytics and Multi-Variant Visualization to Describe Physicochemical Characteristics. Appl. Sci. 2018;8(12):2669. doi: 10.3390/app8122669. [DOI] [Google Scholar]
  801. Schwartz J. B.. The Influence of Sex on Pharmacokinetics. Clinical Pharmacokinetics. 2003;42:107–121. doi: 10.2165/00003088-200342020-00001. [DOI] [PubMed] [Google Scholar]
  802. Chen J., Wang H., Long W., Shen X., Wu D., Song S. S., Sun Y. M., Liu P. X., Fan S., Fan F., Zhang X. D.. Sex Differences in the Toxicity of Polyethylene Glycol-Coated Gold Nanoparticles in Mice. Int. J. Nanomed. 2013;8:2409–2419. doi: 10.2147/IJN.S46376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  803. Zhang X. D., Wu H. Y., Wu D., Wang Y. Y., Chang J. H., Zhai Z., Bin, Meng A. M., Liu P. X., Zhang L. A., Fan F. Y.. Toxicologic Effects of Gold Nanoparticles in Vivo by Different Administration Routes. Int. J. Nanomed. 2010;5(1):771–781. doi: 10.2147/IJN.S8428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  804. Jung T., Kamm W., Breitenbach A., Kaiserling E., Xiao J. X., Kissel T.. Biodegradable Nanoparticles for Oral Delivery of Peptides: Is There a Role for Polymers to Affect Mucosal Uptake? Eur. J. Pharm. Biopharm. 2000;50(1):147–160. doi: 10.1016/S0939-6411(00)00084-9. [DOI] [PubMed] [Google Scholar]
  805. Jia Y. P., Ma B. Y., Wei X. W., Qian Z. Y.. The in Vitro and in Vivo Toxicity of Gold Nanoparticles. Chin. Chem. Lett. 2017;28(4):691–702. doi: 10.1016/j.cclet.2017.01.021. [DOI] [Google Scholar]
  806. Iversen T. G., Skotland T., Sandvig K.. Endocytosis and Intracellular Transport of Nanoparticles: Present Knowledge and Need for Future Studies. Nano Today. 2011;6(2):176–185. doi: 10.1016/j.nantod.2011.02.003. [DOI] [Google Scholar]
  807. Balasubramanian S. K., Jittiwat J., Manikandan J., Ong C. N., Yu L. E., Ong W. Y.. Biodistribution of Gold Nanoparticles and Gene Expression Changes in the Liver and Spleen after Intravenous Administration in Rats. Biomaterials. 2010;31(8):2034–2042. doi: 10.1016/j.biomaterials.2009.11.079. [DOI] [PubMed] [Google Scholar]
  808. Yu M., Zheng J.. Clearance Pathways and Tumor Targeting of Imaging Nanoparticles. ACS Nano. 2015;9:6655–6674. doi: 10.1021/acsnano.5b01320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  809. Perrault S. D., Walkey C., Jennings T., Fischer H. C., Chan W. C. W.. Mediating Tumor Targeting Efficiency of Nanoparticles through Design. Nano Lett. 2009;9(5):1909–1915. doi: 10.1021/nl900031y. [DOI] [PubMed] [Google Scholar]
  810. Mateo D., Paloma M., Alicia Á., Haza A. I.. Oxidative Stress Contributes to Gold Nanoparticle-Induced Cytotoxicity in Human Tumor Cells. Toxicol. Mech. Methods. 2014;24(3):161–172. doi: 10.3109/15376516.2013.869783. [DOI] [PubMed] [Google Scholar]
  811. Fraga S., Faria H., Soares M. E., Duarte J. A., Soares L., Pereira E., Costa-Pereira C., Teixeira J. P., de Lourdes Bastos M., Carmo H.. Influence of the Surface Coating on the Cytotoxicity, Genotoxicity and Uptake of Gold Nanoparticles in Human HepG2 Cells. J. Appl. Toxicol. 2013;33(10):1111–1119. doi: 10.1002/jat.2865. [DOI] [PubMed] [Google Scholar]
  812. Schaeublin N. M., Braydich-Stolle L. K., Schrand A. M., Miller J. M., Hutchison J., Schlager J. J., Hussain S. M.. Surface Charge of Gold Nanoparticles Mediates Mechanism of Toxicity. Nanoscale. 2011;3(2):410–420. doi: 10.1039/c0nr00478b. [DOI] [PubMed] [Google Scholar]
  813. Gao W., Xu K., Ji L., Tang B.. Effect of Gold Nanoparticles on Glutathione Depletion-Induced Hydrogen Peroxide Generation and Apoptosis in HL7702 Cells. Toxicol. Lett. 2011;205(1):86–95. doi: 10.1016/j.toxlet.2011.05.1018. [DOI] [PubMed] [Google Scholar]
  814. Moretti E., Terzuoli G., Renieri T., Iacoponi F., Castellini C., Giordano C., Collodel G.. In Vitro Effect of Gold and Silver Nanoparticles on Human Spermatozoa. Andrologia. 2013;45(6):392–396. doi: 10.1111/and.12028. [DOI] [PubMed] [Google Scholar]
  815. Li J. J., Hartono D., Ong C.-N., Bay B.-H., Yung L.-Y. L.. Autophagy and Oxidative Stress Associated with Gold Nanoparticles. Biomaterials. 2010;31(23):5996–6003. doi: 10.1016/j.biomaterials.2010.04.014. [DOI] [PubMed] [Google Scholar]
  816. Lapresta-Fernández A., Fernández A., Blasco J.. Nanoecotoxicity Effects of Engineered Silver and Gold Nanoparticles in Aquatic Organisms. TrAC, Trends Anal. Chem. 2012;32:40–59. doi: 10.1016/j.trac.2011.09.007. [DOI] [Google Scholar]
  817. Kirubakaran, D. ; Wahid, J. B. A. ; Karmegam, N. ; Jeevika, R. ; Sellapillai, L. ; Rajkumar, M. ; SenthilKumar, K. J. . A Comprehensive Review on the Green Synthesis of Nanoparticles: Advancements in Biomedical and Environmental Applications Biomedical Materials and Devices 2025. 10.1007/s44174-025-00295-4. [DOI]
  818. Bystrzejewska-Piotrowska G., Golimowski J., Urban P. L.. Nanoparticles: Their Potential Toxicity, Waste and Environmental Management. Waste Manage. 2009;29(9):2587–2595. doi: 10.1016/j.wasman.2009.04.001. [DOI] [PubMed] [Google Scholar]
  819. Libralato G., Galdiero E., Falanga A., Carotenuto R., De Alteriis E., Guida M.. Toxicity Effects of Functionalized Quantum Dots, Gold and Polystyrene Nanoparticles on Target Aquatic Biological Models: A Review. Molecules. 2017;22:1439. doi: 10.3390/molecules22091439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  820. Sathya T. A., Viswanathan S., Kolar A. B., Jahirhussain G., Alagumanian S., Sobana S., Arumugam N.. Environmental Profiling of Gold Nanoparticles by Flavonoids Fractionalization from Carrica Papaya Leaf Extract for Photocatalytic Debasement of Organic Contaminants and It’s Cyto-Toxic Analysis. Environ. Res. 2024;259:119445. doi: 10.1016/j.envres.2024.119445. [DOI] [PubMed] [Google Scholar]
  821. Botteon C. E. A., do E S Pereira A., de Castro L. P., Justino I. A., Fraceto L. F., Bastos J. K., Marcato P. D.. Toxicity Assessment of Biogenic Gold Nanoparticles on Crop Seeds and Zebrafish Embryos: Implications for Agricultural and Aquatic Ecosystems. ACS Omega. 2025;10(1):1032–1046. doi: 10.1021/acsomega.4c08287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  822. Jakic K., Selc M., Razga F., Nemethova V., Mazancova P., Havel F., Sramek M., Zarska M., Proska J., Masanova V., Uhnakova I., Makovicky P., Novotova M., Vykoukal V., Babelova A.. Long-Term Accumulation, Biological Effects and Toxicity of BSA-Coated Gold Nanoparticles in the Mouse Liver, Spleen, and Kidneys. Int. J. Nanomedicine. 2024;19:4103–4120. doi: 10.2147/IJN.S443168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  823. Vecchio G., Galeone A., Brunetti V., Maiorano G., Rizzello L., Sabella S., Cingolani R., Pompa P. P.. Mutagenic Effects of Gold Nanoparticles Induce Aberrant Phenotypes in Drosophila Melanogaster. Nanomedicine. 2012;8(1):1–7. doi: 10.1016/j.nano.2011.11.001. [DOI] [PubMed] [Google Scholar]
  824. Al-Shaibani S. W., Alaarage W. K., Wahid M. H. A., Aljdaimi A. I.. Appraisal the Toxicity of CaO and Gold Nanoparticles in Wistar Rats: A Systematic Review. Open Access Res. J. Multidiscip. Stud. 2025;9(1):001–009. doi: 10.53022/oarjms.2025.9.1.0074. [DOI] [Google Scholar]
  825. Mahmoud N. N., Salman T. M., Al-Dabash S., Abdullah M., Abu-Dahab R.. The Impact of Gold Nanoparticles Conjugated with Albumin on Prostate and Breast Cancer Cell Lines: Insights into Cytotoxicity, Cellular Uptake, Migration, and Adhesion Potential. J. Nanopart. Res. 2024;26(5):101. doi: 10.1007/s11051-024-05990-9. [DOI] [Google Scholar]
  826. Kerdtoob S., Chanthasena P., Rosyidah A., Limphirat W., Penkhrue W., Ganta P., Srisakvarangkool W., Yasawong M., Nantapong N.. Streptomyces Monashensis MSK03-Mediated Synthesis of Gold Nanoparticles: Characterization and Antibacterial Activity. RSC Adv. 2024;14(7):4778–4787. doi: 10.1039/D3RA07555A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  827. Kim S., Wang R., Dhandapani S., Kang K., Cho I. H., Kim Y. J.. Novel Modified Probiotic Gold Nanoparticles Loaded with Ginsenoside CK Exerts an Anti-Inflammation Effect via NF-KB/MAPK Signaling Pathways. Arabian J. Chem. 2024;17(4):105650. doi: 10.1016/j.arabjc.2024.105650. [DOI] [Google Scholar]
  828. Aljohani F. S., Hamed M. T., Bakr B. A., Shahin Y. H., Abu-Serie M. M., Awaad A. K., El-Kady H., Elwakil B. H.. In Vivo Bio-Distribution and Acute Toxicity Evaluation of Greenly Synthesized Ultra-Small Gold Nanoparticles with Different Biological Activities. Sci. Rep. 2022;12(1):6269. doi: 10.1038/s41598-022-10251-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  829. Vetten M. A., Tlotleng N., Rascher D. T., Skepu A., Keter F. K., Boodhia K., Koekemoer L. A., Andraos C., Tshikhudo R., Gulumian M.. Label-Free in Vitro Toxicity and Uptake Assessment of Citrate Stabilised Gold Nanoparticles in Three Cell Lines. Part. Fibre Toxicol. 2013;10(1):50. doi: 10.1186/1743-8977-10-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  830. Stelzer R., Hutz R. J.. Gold Nanoparticles Enter Rat Ovarian Granulosa Cells and Subcellular Organelles, and Alter In-Vitro Estrogen Accumulation. J. Reproduct. Dev. 2009;55:685–690. doi: 10.1262/jrd.20241. [DOI] [PubMed] [Google Scholar]
  831. Raji V., Kumar J., Rejiya C. S., Vibin M., John A., Abraham A.. Synthesis, Characterisation and Biocompatibility of Surface-Functionalised Gold Nanoparticles. J. Exp. Nanosci. 2012;7(2):174–188. doi: 10.1080/17458080.2010.514952. [DOI] [Google Scholar]
  832. Leifert A., Pan Y., Kinkeldey A., Schiefer F., Setzler J., Scheel O., Lichtenbeld H., Schmid G., Wenzel W., Jahnen-Dechent W., Simon U.. Differential HERG Ion Channel Activity of Ultrasmall Gold Nanoparticles. Proc. Natl. Acad. Sci. U.S.A. 2013;110(20):8004–8009. doi: 10.1073/pnas.1220143110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  833. Das S., Debnath N., Mitra S., Datta A., Goswami A.. Comparative Analysis of Stability and Toxicity Profile of Three Differently Capped Gold Nanoparticles for Biomedical Usage. BioMetals. 2012;25(5):1009–1022. doi: 10.1007/s10534-012-9567-1. [DOI] [PubMed] [Google Scholar]
  834. Schneider T., Westermann M., Glei M.. In Vitro Uptake and Toxicity Studies of Metal Nanoparticles and Metal Oxide Nanoparticles in Human HT29 Cells. Arch. Toxicol. 2017;91(11):3517–3527. doi: 10.1007/s00204-017-1976-z. [DOI] [PubMed] [Google Scholar]
  835. Coradeghini R., Gioria S., García C. P., Nativo P., Franchini F., Gilliland D., Ponti J., Rossi F.. Size-Dependent Toxicity and Cell Interaction Mechanisms of Gold Nanoparticles on Mouse Fibroblasts. Toxicol. Lett. 2013;217(3):205–216. doi: 10.1016/j.toxlet.2012.11.022. [DOI] [PubMed] [Google Scholar]
  836. Paino I. M. M., Marangoni V. S., de Cássia Silva de Oliveira R., Antunes L. M. G., Zucolotto V.. Cyto and Genotoxicity of Gold Nanoparticles in Human Hepatocellular Carcinoma and Peripheral Blood Mononuclear Cells. Toxicol. Lett. 2012;215(2):119–125. doi: 10.1016/j.toxlet.2012.09.025. [DOI] [PubMed] [Google Scholar]
  837. Reipa V., Hackley V. A., Tona A., Heo M. B., Lee Y. R., Lee T. G., Johnston-Peck A., Cho T. J.. Well-Characterized Polyethyleneimine-/Carboxylated-Polyethylene-Glycol-Functionalized Gold Nanoparticles as Prospective Nanoscale Control Materials for In Vitro Cell Viability Assays: Particle Characterization and Toxicity Tests in Eight Mammalian Cell Lines. Nanomaterials. 2025;15(2):79. doi: 10.3390/nano15020079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  838. Salado J., Insausti M., Lezama L., Gil De Muro I., Moros M., Pelaz B., Grazu V., De La Fuente J. M., Rojo T.. Functionalized Fe 3 O 4 @Au Superparamagnetic Nanoparticles: In Vitro Bioactivity. Nanotechnology. 2012;23:315102. doi: 10.1088/0957-4484/23/31/315102. [DOI] [PubMed] [Google Scholar]
  839. Karbalaee M., Sadeghi Z., Ahmadianpour M. V., Jahangiri B., Valipour R., Aleyasin S. A., Raheb J.. Nanoparticle-Protein Corona Interactions in Biological Milieu: Differential Toxicity Profiles in Prostate Cancer and Normal Cell Lines. J. Sci. Islamic Republic of Iran. 2024;35(2):115–124. [Google Scholar]
  840. Schaeublin N. M., Braydich-Stolle L. K., Schrand A. M., Miller J. M., Hutchison J., Schlager J. J., Hussain S. M.. Surface Charge of Gold Nanoparticles Mediates Mechanism of Toxicity. Nanoscale. 2011;3(2):410–420. doi: 10.1039/c0nr00478b. [DOI] [PubMed] [Google Scholar]
  841. Tedesco S., Doyle H., Blasco J., Redmond G., Sheehan D.. Oxidative Stress and Toxicity of Gold Nanoparticles in Mytilus Edulis. Aquat. Toxicol. 2010;100(2):178–186. doi: 10.1016/j.aquatox.2010.03.001. [DOI] [PubMed] [Google Scholar]
  842. Fraga S., Brandão A., Soares M. E., Morais T., Duarte J. A., Pereira L., Soares L., Neves C., Pereira E., de Lourdes Bastos M., Carmo H.. Short- and Long-Term Distribution and Toxicity of Gold Nanoparticles in the Rat after a Single-Dose Intravenous Administration. Nanomedicine. 2014;10(8):1757–1766. doi: 10.1016/j.nano.2014.06.005. [DOI] [PubMed] [Google Scholar]
  843. Sabo-Attwood T., Unrine J. M., Stone J. W., Murphy C. J., Ghoshroy S., Blom D., Bertsch P. M., Newman L. A.. Uptake, Distribution and Toxicity of Gold Nanoparticles in Tobacco (Nicotiana Xanthi) Seedlings. Nanotoxicology. 2012;6(4):353–360. doi: 10.3109/17435390.2011.579631. [DOI] [PubMed] [Google Scholar]
  844. Bozich J. S., Lohse S. E., Torelli M. D., Murphy C. J., Hamers R. J., Klaper R. D.. Surface Chemistry, Charge and Ligand Type Impact the Toxicity of Gold Nanoparticles to Daphnia Magna. Environ. Sci. Nano. 2014;1(3):260–270. doi: 10.1039/C4EN00006D. [DOI] [Google Scholar]
  845. Simpson C. A., Salleng K. J., Cliffel D. E., Feldheim D. L.. In Vivo Toxicity, Biodistribution, and Clearance of Glutathione-Coated Gold Nanoparticles. Nanomedicine. 2013;9(2):257–263. doi: 10.1016/j.nano.2012.06.002. [DOI] [PubMed] [Google Scholar]
  846. Thakor A. S., Luong R., Paulmurugan R., Lin F. I., Kempen P., Zavaleta C., Chu P., Massoud T. F., Sinclair R., Gambhir S. S.. The Fate and Toxicity of Raman-Active Silica-Gold Nanoparticles in Mice. Sci. Transl Med. 2011;3(79):79ra33. doi: 10.1126/scitranslmed.3001963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  847. Vujačić A., Petrovic S., Vodnik V., Joksić G., Petrović S., Leskovac A., Nastasijević B., Vasić V.. Particle Size and Concentration Dependent Cytotoxicity of Citrate Capped Gold Nanoparticles. Digest J. Nanomater. Biostruct. 2011;6:1367–1376. [Google Scholar]
  848. Sereemaspun, A. ; Pongsuchart, M. ; Danladkaew, C. ; Khomvarn, T. In Effect of Glutathione-Stabilized Gold Nanoparticles in 3T3 Fibroblast Cell, 2012 International Conference on Clean and Green Energy; IACSIT Press, 2012. [Google Scholar]
  849. Kus-liśkiewicz M., Fickers P., Ben Tahar I.. Biocompatibility and Cytotoxicity of Gold Nanoparticles: Recent Advances in Methodologies and Regulations. Int. J. Mol. Sci. 2021;22:10952. doi: 10.3390/ijms222010952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  850. Wacker M. G., Proykova A., Santos G. M. L.. Dealing with Nanosafety around the GlobeRegulation vs. Innovation. Int. J. Pharm. 2016;509(1):95–106. doi: 10.1016/j.ijpharm.2016.05.015. [DOI] [PubMed] [Google Scholar]
  851. Council, N. R. Review of the Federal Strategy for Nanotechnology-Related Environmental, Health, and Safety Research; The National Academies Press: Washington, DC, 2009. 10.17226/12559. [DOI] [PubMed] [Google Scholar]
  852. Contains Nonbinding Recommendations Guidance for Industry Assessing the Effects of Significant Manufacturing Process Changes, Including Emerging Technologies, on the Safety and Regulatory Status of Food Ingredients and Food Contact Substances, Including Food Ingredients That Are Color Additives. http://www.regulations.gov.
  853. Bobo D., Robinson K. J., Islam J., Thurecht K. J., Corrie S. R.. Nanoparticle-Based Medicines: A Review of FDA-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016;33(10):2373–2387. doi: 10.1007/s11095-016-1958-5. [DOI] [PubMed] [Google Scholar]
  854. Sintes, J. R. ; Roebben, G. ; Linsinger, T. P. J. ; Gibson, N. ; Rauscher, H. ; Kestens, V. ; Mech, A. . An Overview of Concepts and Terms Used in the European Commission’s Definition of Nanomaterial; Publications Office of the European Union, 2019. [Google Scholar]
  855. Human Health and Environmental Exposure Assessment and Risk Characterisation of Nanomaterials – Best Practice for REACH Registrants – Third GAARN Meeting, Helsinki, 30 September 2013; European Chemicals Agency, 2014. [Google Scholar]
  856. Guidance on the Determination of Potential Health Effects of Nanomaterials Used in Medical Devices; European Commission, 2015. [Google Scholar]
  857. Bernauer U., Bodin L., Chaudhry Q., Coenraads P. J., Dusinska M., Gaffet E., Panteri E., Rogiers V., Rousselle C., Stepnik M., Vanhaecke T., Wijnhoven S., von Goetz N., de Jong W. H., Simonnard A.. The SCCS Guidance on the Safety Assessment of Nanomaterials in Cosmetics. Regul. Toxicol. Pharmacol. 2020;112:104611. doi: 10.1016/j.yrtph.2020.104611. [DOI] [PubMed] [Google Scholar]
  858. Schoonjans R., Tarazona J.. European Food Safety Authority (EFSA) Annual Report of the EFSA Scientific Network of Risk Assessment of Nanotechnologies in Food and Feed for 2020. EFSA Supporting Publ. 2021;18(3):6502E. doi: 10.2903/sp.efsa.2021.EN-6502. [DOI] [Google Scholar]
  859. European Medicines Agency . Committee for Medicinal Products for Human Use (CHMP) Reflection Paper on Surface Coatings: General Issues for Consideration Regarding Parenteral Administration of Coated Nanomedicine Products Reflection Paper on Surface Coatings: General Issues for Consideration Regarding Parenteral Administration of Coated Nanomedicine Products. www.ema.europa.eu.
  860. Contains Nonbinding Recommendations 1 Guidance for Industry Safety of Nanomaterials in Cosmetic Products. http://www.regulations.gov.
  861. Yadav S., Mali S. N., Pandey A.. Biogenic Nanoparticles as Safer Alternatives for Gastric Ulcers: An Update on Green Synthesis Methods, Toxicity, and Their Efficacy in Controlling Inflammation. Biol. Trace Elem Res. 2025;203:3967–3986. doi: 10.1007/s12011-024-04446-4. [DOI] [PubMed] [Google Scholar]
  862. Das U., Mali S. N., Mandal S.. Unveiling the Ameliorative Effects of Phyto-Selenium Nanoparticles (PSeNPs) on Anti-Hyperglycemic Activity and Hyperglycemia Irradiated Complications. BioNanoScience. 2025;15(2):231. doi: 10.1007/s12668-025-01830-9. [DOI] [Google Scholar]
  863. Dheyab M. A., Aziz A. A., Jameel M. S., Khaniabadi P. M.. Recent Advances in Synthesis, Medical Applications and Challenges for Gold-Coated Iron Oxide: Comprehensive Study. Nanomaterials. 2021;11:2147. doi: 10.3390/nano11082147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  864. Ali Dheyab, M. ; Tang, J. H. ; Abdul Aziz, A. ; Hussein Nowfal, S. ; Jameel, M. S. ; Alrosan, M. ; Oladzadabbasabadi, N. ; Ghasemlou, M. . Green Synthesis of Gold Nanoparticles and Their Emerging Applications in Cancer Imaging and Therapy: A Review. 2024. 10.1515/revic-2024-0048. [DOI]
  865. Dheyab M. A., Aziz A. A., Khaniabadi P. M., Jameel M. S., Oladzadabbasabadi N., Rahman A. A., Braim F. S., Mehrdel B.. Gold Nanoparticles-Based Photothermal Therapy for Breast Cancer. Photodiagn. Photodyn. Ther. 2023;42:103312. doi: 10.1016/j.pdpdt.2023.103312. [DOI] [PubMed] [Google Scholar]
  866. Ali Dheyab M., Abdul Aziz A., Jameel M. S., Moradi Khaniabadi P., Mehrdel B.. Sonochemical-Assisted Synthesis of Highly Stable Gold Nanoparticles Catalyst for Decoloration of Methylene Blue Dye. Inorg. Chem. Commun. 2021;127:108551. doi: 10.1016/j.inoche.2021.108551. [DOI] [Google Scholar]
  867. Ali Dheyab M., Abdul Aziz A., Moradi Khaniabadi P., Jameel M. S., Ahmed N. M., Taha Ali A.. Distinct Advantages of Using Sonochemical over Laser Ablation Methods for a Rapid-High Quality Gold Nanoparticles Production. Mater. Res. Express. 2021;8(1):015009. doi: 10.1088/2053-1591/abd5a4. [DOI] [Google Scholar]
  868. Dheyab M. A., Aziz A. A., Jameel M. S.. Synthesis and Optimization of the Sonochemical Method for Functionalizing Gold Shell on Fe3O4 Core Nanoparticles Using Response Surface Methodology. Surf. Interfaces. 2020;21:100647. doi: 10.1016/j.surfin.2020.100647. [DOI] [Google Scholar]
  869. Dheyab M. A., Aziz A. A., Oladzadabbasabadi N., Alsaedi A., Braim F. S., Jameel M. S., Ramizy A., Alrosan M., Almajwal A. M.. Comparative Analysis of Stable Gold Nanoparticles Synthesized Using Sonochemical and Reduction Methods for Antibacterial Activity. Molecules. 2023;28(9):3931. doi: 10.3390/molecules28093931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  870. Dheyab M. A., Aziz A. A., Jameel M. S., Khaniabadi P. M., Oglat A. A.. Rapid Sonochemically-Assisted Synthesis of Highly Stable Gold Nanoparticles as Computed Tomography Contrast Agents. Appl. Sci. 2020;10(20):7020. doi: 10.3390/app10207020. [DOI] [Google Scholar]
  871. Dheyab M. A., Aziz A. A., Jameel M. S., Khaniabadi P. M., Mehrdel B.. Mechanisms of Effective Gold Shell on Fe3O4 Core Nanoparticles Formation Using Sonochemistry Method. Ultrason. Sonochem. 2020;64:104865. doi: 10.1016/j.ultsonch.2019.104865. [DOI] [PubMed] [Google Scholar]
  872. Dheyab M. A., Aziz A. A., Jameel M. S., Noqta O. A., Khaniabadi P. M., Mehrdel B.. Excellent Relaxivity and X-Ray Attenuation Combo Properties of Fe3O4@Au CSNPs Produced via Rapid Sonochemical Synthesis for MRI and CT Imaging. Mater. Today Commun. 2020;25:101368. doi: 10.1016/j.mtcomm.2020.101368. [DOI] [Google Scholar]
  873. Dheyab M. A., Owaid M. N., Rabeea M. A., Aziz A. A., Jameel M. S.. Mycosynthesis of Gold Nanoparticles by the Portabello Mushroom Extract, Agaricaceae, and Their Efficacy for Decolorization of Azo Dye. Environ. Nanotechnol. Monit. Manage. 2020;14:100312. doi: 10.1016/j.enmm.2020.100312. [DOI] [Google Scholar]
  874. Dheyab M. A., Aziz A. A., Al-Mafarjy S. S., Suardi N., Razak N. N. A. N. A., Ramizy A., Jameel M. S.. Exploring the Anticancer Potential of Biogenic Inorganic Gold Nanoparticles Synthesized via Mushroom-Assisted Green Route. Inorg. Chem. Commun. 2023;157:111363. doi: 10.1016/j.inoche.2023.111363. [DOI] [Google Scholar]
  875. Owaid M. N., Rabeea M. A., Abdul Aziz A., Jameel M. S., Dheyab M. A.. Mushroom-Assisted Synthesis of Triangle Gold Nanoparticles Using the Aqueous Extract of Fresh Lentinula Edodes (Shiitake), Omphalotaceae. Environ. Nanotechnol. Monit. Manage. 2019;12:100270. doi: 10.1016/j.enmm.2019.100270. [DOI] [Google Scholar]
  876. Rabeea M. A., Owaid M. N., Aziz A. A., Jameel M. S., Dheyab M. A.. Mycosynthesis of Gold Nanoparticles Using the Extract of Flammulina Velutipes, Physalacriaceae, and Their Efficacy for Decolorization of Methylene Blue. J. Environ. Chem. Eng. 2020;8(3):103841. doi: 10.1016/j.jece.2020.103841. [DOI] [Google Scholar]
  877. Dheyab M. A., Aziz A. A., Khaniabadi P. M., Jameel M. S., Oladzadabbasabadi N., Mohammed S. A., Abdullah R. S., Mehrdel B.. Monodisperse Gold Nanoparticles: A Review on Synthesis and Their Application in Modern Medicine. Int. J. Mol. Sci. 2022;23:7400. doi: 10.3390/ijms23137400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  878. Dheyab M. A., Abdul Aziz A., Hussein Nowfal S., Al-Mafarjy S. S., Abdullah W., Suardi N., Jameel M. S., Braim F. S., Alrosan M., Khaniabadi P. M.. Turning Food Waste-Derived Ultrasmall Gold Nanoparticles as a Photothermal Agent for Breast Cancer Cell Eradication. Inorg. Chem. Commun. 2024;169:113030. doi: 10.1016/j.inoche.2024.113030. [DOI] [Google Scholar]
  879. Dhawan, S. ; Sharma, P. ; Nanda, S. . Cosmetic Nanoformulations and Their Intended Use. In Nanocosmetics; Nanda, A. ; Nanda, S. ; Nguyen, T. A. ; Rajendran, S. ; Slimani, Y. , Eds.; Elsevier, 2020; Chapter 8, pp 141–169 10.1016/B978-0-12-822286-7.00017-6. [DOI] [Google Scholar]
  880. Dubey S. K., Dey A., Singhvi G., Pandey M. M., Singh V., Kesharwani P.. Emerging Trends of Nanotechnology in Advanced Cosmetics. Colloids Surf., B. 2022;214:112440. doi: 10.1016/j.colsurfb.2022.112440. [DOI] [PubMed] [Google Scholar]
  881. Alharthi S. D., Bijukumar D., Prasad S., Khan A. M., Mathew M. T.. Evolution in Biosensors for Cancers Biomarkers Detection: A Review. J. Bio- Tribo-Corros. 2021;7(2):42. doi: 10.1007/s40735-020-00463-7. [DOI] [Google Scholar]
  882. Fox J., Newham G., Bushby R. J., Valleley E. M. A., Coletta P. L., Evans S. D.. Spectrophotometric Analysis and Optimization of 2D Gold Nanosheet Formation. J. Phys. Chem. C. 2023;127(6):3067–3076. doi: 10.1021/acs.jpcc.2c07582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  883. Huang W., Xu Y., Wang Z., Liao K., Zhang Y., Sun Y.. Dual Nanozyme Based on Ultrathin 2D Conductive MOF Nanosheets Intergraded with Gold Nanoparticles for Electrochemical Biosensing of H2O2 in Cancer Cells. Talanta. 2022;249:123612. doi: 10.1016/j.talanta.2022.123612. [DOI] [PubMed] [Google Scholar]
  884. Rauf S., Lahcen A. A., Aljedaibi A., Beduk T., Ilton de Oliveira Filho J., Salama K. N.. Gold Nanostructured Laser-Scribed Graphene: A New Electrochemical Biosensing Platform for Potential Point-of-Care Testing of Disease Biomarkers. Biosens. Bioelectron. 2021;180:113116. doi: 10.1016/j.bios.2021.113116. [DOI] [PubMed] [Google Scholar]
  885. Mohammadniaei M., Koyappayil A., Sun Y., Min J., Lee M.-H.. Gold Nanoparticle/MXene for Multiple and Sensitive Detection of OncomiRs Based on Synergetic Signal Amplification. Biosens. Bioelectron. 2020;159:112208. doi: 10.1016/j.bios.2020.112208. [DOI] [PubMed] [Google Scholar]
  886. Abad A., Concepción P., Corma A., García H.. A Collaborative Effect between Gold and a Support Induces the Selective Oxidation of Alcohols. Angew. Chem., Int. Ed. 2005;44(26):4066–4069. doi: 10.1002/anie.200500382. [DOI] [PubMed] [Google Scholar]
  887. Xiao L., Zhu A., Xu Q., Chen Y., Xu J., Weng J.. Colorimetric Biosensor for Detection of Cancer Biomarker by Au Nanoparticle-Decorated Bi2Se3 Nanosheets. ACS Appl. Mater. Interfaces. 2017;9(8):6931–6940. doi: 10.1021/acsami.6b15750. [DOI] [PubMed] [Google Scholar]
  888. Selwyna, P. G. C. ; Loganathan, P. R. ; Begam, K. H. In Development of Electrochemical Biosensor for Breast Cancer Detection Using Gold Nanoparticle Doped CA 15–3 Antibody and Antigen Interaction, 2013 International Conference on Signal Processing, Image Processing & Pattern Recognition; IEEE, 2013; pp 75–81. [Google Scholar]
  889. Ranjan P., Abubakar Sadique M., Yadav S., Khan R.. An Electrochemical Immunosensor Based on Gold-Graphene Oxide Nanocomposites with Ionic Liquid for Detecting the Breast Cancer CD44 Biomarker. ACS Appl. Mater. Interfaces. 2022;14(18):20802–20812. doi: 10.1021/acsami.2c03905. [DOI] [PubMed] [Google Scholar]

Articles from ACS Omega are provided here courtesy of American Chemical Society

RESOURCES