Abstract

Amphiphilic macrocycles, such as p-sulfonatocalix[6]arenes (p-SC6), have demonstrated great potential in designing synthetic nanovesicles based on self-assembly approaches. These supramolecular nanovesicles are capable of improving the solubility, stability, and biological activity of various drugs. In the present study, the biologically active harmala alkaloid-rich fraction (HARF) was extracted from Peganum harmala L. seeds. Ultraperformance liquid chromatography–electrospray ionization–tandem mass spectrometry (UPLC/ESI-MS) analysis of HARF revealed 15 alkaloids. The reversed-phase high-performance liquid chromatography (RP-HPLC) analysis revealed three peaks: peganine, harmol, and harmine. The HARF was then encapsulated in p-SC6 nanocapsules employing a thin-film hydration approach. The designed nanocapsules had an average particle size of 264.8 ± 10.6 nm, and a surface charge of −30.3 ± 2.2 mV. They were able to encapsulate 89.3 ± 1.4, 74.4 ± 1.3, and 76.1 ± 1.7% of the three harmala alkaloids; harmine, harmol, and peganine; respectively. The in vitro drug release experiments showed the potential of the designed nanocapsules to release their cargo at a pH of 5.5 (typical of cancerous tissue). The IC50 values of HARF encapsulated in p-SC6 (H/p-SC6 nanocapsules) were 5 and 2.7 μg/mL against ovarian cancer cells (SKOV-3) and breast adenocarcinoma cells (MCF-7), respectively. The prepared nanocapsules were found to be biocompatible when tested on human skin fibroblasts. Additionally, the antioxidant activity of the designed nanocapsules was 5 times that of the free powder fraction; the IC50 of the H/p-SC6 nanocapsules was 30.1 ± 1.3 μg/mL, and that of the HARF was 169.3 ± 7.2 μg/mL. In conclusion, encapsulation of P. harmala alkaloid-rich fraction into self-assembled p-SC6 significantly increases its antioxidant and cytotoxic activities.
1. Introduction
Cancer is the second most common cause of death in developed and developing countries, with a steady increase in the number of new cases each year. The number of patients diagnosed with cancer is predicted to increase by 2-fold within the coming 30 years.1 Currently, synthetic chemotherapeutic agents are used in cancer therapy either alone or in combination with surgical resection and/or radiation therapy.2−7 However, direct use of classical chemotherapeutic agents and procedures has many drawbacks and generates severe adverse effects and resistance.8−10 However, encapsulation of the anticancer drugs into nanocarriers could provide effective drug delivery and minimize their side effects.11−13 Since more than 50% of the currently available anticancer drugs are from natural products or developed derivatives, natural products should be recognized as a potential source of potent and less toxic anticancer agents. Many studies have been reported on this path on employing natural products to treat several diseases.14,15
Peganum harmala L. (Zygophyllaceae) is a small herbaceous plant known to grow as an invasive weed on poor soils in the Middle East and North Africa.16 The mature harmala seeds are rich in alkaloids (about 6% of total dry weight), including some β-carboline and quinazoline alkaloids, jointly known as harmala alkaloids.17 Major harmala alkaloids include harmine, harmane, harmol, harmaline, harmalol, and peganine. These alkaloids possess a broad spectrum of biological activities, including antibacterial, antioxidant, antiplatelet, immunomodulatory, and cytotoxic activities.16,18 However, their clinical applications in cancer therapy are hindered by their hydrophobic nature,16 and some studies reported toxic effects of harmala alkaloids on the central nervous system at high doses.19 Accordingly, carrying out encapsulation of harmala alkaloids could effectively reduce their harmful effects and/or enhance their anticancer activities.
Various nanocarriers such as polymeric nanoparticles, dendrimers, and liposomes were previously reported for the targeted delivery of different natural and synthetic drugs.15,20 Recently, macromolecules, such as p-sulfonato-calix[n]arenes [n = 4 and 6, (p-SC4 and p-SC6)] have demonstrated considerable impact on the delivery of anticancer drugs.10p-SCns are water-soluble, safe (for doses up to 105 μg/kg), biodegradable, and biocompatible. They are very promising as a potential host molecule for various chemotherapeutics via host–guest inclusion complexation.21−24 Additionally, amphiphilic macrocycles can be used to develop nanocapsules relying on self-assembly methods. These assemblies are bonded together via weak and reversible bonds, making them very responsive. Therefore, their amphiphilic nature qualifies them to encapsulate both water-soluble and oil-soluble chemotherapeutic agents within their central shells where host–guest complexation plays a role part in binding the hydrophobic and hydrophilic moieties.21,25 These assemblies were also reported to exhibit targeted and controlled release of drugs to the sites of action and hence minimize the undesirable effects on healthy cells. The first amphiphilic macrocyclic nanocapsules based on self-assembly advances were designed by Markowitz et al. (1989) by injecting tetrahydrofuran solution containing p-SC6 into water resulting in the production of nanocapsules with size ranging from 500 to 1000 nm.26 Further studies have been conducted on the design of p-SCn as potential drug carriers. For example, nanovesicles created by the self-assembly of tetrahexyloxy-p-sulfonatocalix[4]arene were utilized to encapsulate paclitaxel (PTX) anticancer drug. The prepared nanocapsules exhibited much enhanced anticancer activities on human cervical cancer cells than the unencapsulated PTX, at concentrations of 1, 10, and 100 μg/mL.27
In the present work, the harmala alkaloid-rich fraction extracted from P. harmala L. seeds was encapsulated in p-SC6 nanocapsules (H/p-SC6) employing a thin-film hydration approach. Nanocapsules were physically identified in terms of average particle sizes, surface charge, polydispersity index (PDI), encapsulation efficiency %, and morphology (employing transmission electron microscope (TEM)). Also, the chemical structural features of the nanocapsules were investigated employing Fourier transform infrared spectroscopy (FT-IR). Additionally, the antioxidant (using DPPH) and cytotoxic activities against ovarian cancer cells (SKOV-3), breast adenocarcinoma (MCF-7), and human skin fibroblasts (using sulforhodamine B (SRB) assay) were evaluated for both encapsulated and unencapsulated harmala alkaloid-rich fraction.
2. Results and Discussion
2.1. P. harmala Alkaloids
The three major alkaloids of P. harmala (peganine, harmol, and harmine) were detected using HPLC–DAD and confirmed by UPLC-ESI-MS analysis.
2.1.1. Detection of Harmala Alkaloids by HPLC–DAD
The alkaloids of P. harmala seeds were analyzed on HPLC–DAD. The chromatogram showed three major alkaloids, which could be detected by HPLC–DAD at 320 nm and corresponded to peganine, harmol, and harmine (Figure 1) as previously described.16 These peaks were compared to standard harmine and the isolated alkaloids peganine, harmol, and harmine. The isolated ones were annotated by their mass and UV spectra (Figures S1 and S2). Moreover, these alkaloids, in addition to the other 15 alkaloids, were confirmed by liquid chromatography with tandem mass spectrometry (LC-MS/MS).
Figure 1.
HPLC chromatogram of P. harmala seed total alkaloid extract using a Waters LC 2695 coupled to a Waters 996 diode array detector (DAD) at 320 nm. XTerra MS-C18 column (100 × 4.6 mm2 id, 5 μm). Mobile phase: water containing triethylamine (pH 8.6) (A) and acetonitrile (B). Gradient elution from 0% B (100% A) to 20% B in 4 min, 30% B at 9 min, 50% B at 14 min, and 60% B at 16 min. Then, 2 min washing with 100% B and reequilibration for 4 min. A flow rate of 1 mL/min, column temperature of 25 °C, and injection volume of 20 μL were applied. The peaks were compared to standard harmine and the isolated alkaloids (peganine, harmol, and harmine).
2.1.2. Analysis of Harmala Alkaloid-Rich Fraction by Ultraperformance Liquid Chromatography–Electrospray Ionization–Tandem Mass Spectrometry (UPLC/ESI-MS)
The phytochemical identification of the harmala alkaloid-rich fraction (HARF) was performed using UPLC-ESI-MS analysis. Fifteen alkaloids were timidly detected using their m/z, fragmentation pattern, their elution order from the HPLC column, and by comparing their mass spectral data with those reported. The annotated alkaloids are either β-carboline or quinazoline-type alkaloids. Alkaloids of both types are major compounds in this plant.28 The major β-carboline alkaloids annotated were harmine (m/z [M + H]+ 213.1546) and harmaline (m/z [M + H]+ 215.5396), with MS2 product patterns at 198 and 170 (for harmine) and 200 and 174 (for harmaline). However, quinazoline alkaloids were represented mainly by vasicine (peganine) and vasicinone (m/z [M + H]+ 189.2218 and 203.1254; MS2 ions at 171, 154, 144, and 117 “for vasicine”, and 203 and 185 “for vasicinone”). Other perceived major harmala alkaloids were pegamine, hydroxylated harmine, and tetrahydroharmine. In addition, some minor alkaloids such as vasicinone hexoside, harmalol, deoxyvasicinone, hydroxylated harmaline, harmol, and harmalanine are presented in Table 1 and Figure 2. The structures of the major detected alkaloids are depicted in Figure 3, and the MS2 fragmentation pattern of selected alkaloids is represented in Figure S3.
Table 1. Tentatively Identified Peaks in the LC-MS Spectrum of the Total Alkaloids Fraction of P. harmala Seed Extract.
| peak no. | assignment | formula | RT (min) | m/z [M + H]+ | product ions MS/MS | refs |
|---|---|---|---|---|---|---|
| 1 | vasicine (peganine) | C11H13N2O+ | 0.79 | 189.3991 | 189, 171, 154, 144, 130, 117 | (29, 30) |
| 2 | vasicine (peganine) isomer | C11H13N2O+ | 1.20 | 189.2218 | 189, 171, 154, 144, 117 | (29−31) |
| 3 | vasicinone hexoside | C17H21N2O7+ | 2.06 | 365.1631 | 365, 203, 185, 82 | (32) |
| 4 | harmalol | C12H13N2O+ | 2.18 | 201.1134 | 201, 184, 160, 67 | (29, 30) |
| 5 | pegamine | C11H13N2O2+ | 2.31 | 205.1031 | 187, 120, 74 | (29, 33) |
| 6 | vasicinone | C11H11N2O2+ | 2.62 | 203.1254 | 203, 185, 82 | (29, 30) |
| 7 | hydroxylated harmine | C13H13N2O2+ | 4.00 | 229.1012 | 229, 214, 186, 211 | (29, 30) |
| 8 | deoxyvasicinone | C11H11N2O+ | 4.56 | 187.0925 | 187, 159, 144, 131 | (30) |
| 9 | tetrahydroharmine | C13H17N2O+ | 4.83 | 217.1005 | 217, 199.9, 188, 173.9, 169, 157 | (29, 30) |
| 10 | harmaline | C13H15N2O+ | 5.40 | 215.5396 | 215, 200,174 | (29, 30) |
| 11 | harmine | C13H13N2O+ | 5.81 | 213.1546 | 198,170 | (29, 30) |
| 12 | hydroxylated harmaline | C13H15N2O2+ | 6.60 | 231.1150 | 214, 199, 187 | (34) |
| 13 | unknown | 7.17 | 2. 2321 | 213, 196, 188, 160 | (28) | |
| 14 | tetrahydroharmine isomer | C13H17N2O+ | 7.77 | 217.1005 | 217, 199.9, 188, 173.9, 169, 157 | (29, 30) |
| 15 | harmol | C12H11N2O+ | 8.43 | 199.1732 | 199, 181, 171, 131, 105, 73 | (29, 35) |
| 16 | unknown | 9.24 | 379.2151 | 364, 335 | ||
| 17 | unknown | 9.82 | 441.2976 | 268, 215, 201, 174 | ||
| 18 | unknown | 10.49 | 481.3096 | |||
| 19 | unknown | 11.81 | 274.2809 | |||
| 20 | hexadecenal | C16H31O+ | 20.76 | 239.2334 | 239, 221, 195, 169, 155, 141, 113, 99, 85, 71, 57, 43 | (36) |
| 21 | harmalanine | C16H15N2O2+ | 23.48 | 267.2690 | 253, 236, 206, 186 | (37) |
| 22 | unknown | 25.11 | 413.3079 | 268, 215, 174 |
Figure 2.
UPLC-ESI-MS/MS chromatogram (in positive-ion mode) of the total alkaloids fraction of P. harmala separated on an ACQUITY UPLC-BEH C18 (1.7 μm, 2.1 × 50 mm2) column.
Figure 3.

Major alkaloids detected in HARF by ultraperformance liquid chromatography–electrospray ionization–tandem mass spectrometry (UPLC/ESI-MS).
2.2. Identification of the Designed Harmala Alkaloid-Rich Fraction/p-SC6 (H/p-SC6) Supramolecular Nanocapsules
2.2.1. Particle Size, Polydispersity Index (PDI), ζ-Potential, and Encapsulation Efficiency (EE)
The average size and PDI of the designed H/p-SC6 system were investigated by dynamic light scattering and were found to be 264.77 ± 10.56 nm and 0.26 ± 0.02, respectively (Table 2). These values lie within the range of the particles with nano sizes of 200–600 nm previously reported as ideal chemotherapeutics carriers with boosted passive accumulation, due to enhanced permeability and retention (EPR), into the permeable vasculature of tumor cells.38−40 The ζ-potential of the designed nanocapsules was obtained via laser Doppler velocimetry and exhibited a high negative surface charge of −30.33 ± 2.15 mV. This is attributed to the presence of the anionic p-SC6 amphiphile, which is highly negatively charged.21 The increased surface negativity on the nanocapsules is expected to impart high stability as it inhibits the aggregation of the particles, and hence keeps them suspended for a longer period. Encapsulation efficiencies on the three alkaloids are presented in Table 2.
Table 2. Average Size, PDI, ζ-Potential, and Encapsulation Efficiency of H/p-SC6 Nanocapsules.
| encapsulation efficiency (%) ± SD |
||||||
|---|---|---|---|---|---|---|
| formula | average size (nm) | PDI | ζ-potential (mV) ± SD | harmine | harmol | peganine |
| H/p-SC6 | 264.77 ± 10.56 | 0.26 ± 0.02 | –30.33 ± 2.15 | 89.34 ± 1.41 | 74.39 ± 1.30 | 76.10 ± 1.66 |
The high encapsulation efficiency of HARF is attributed to the unique chemical structure of the amphiphilic p-SC6 that possesses a para-substituent of a phenolic ring on the upper edge, a phenolic hydroxyl group on the lower edge, and a hydrophobic π electron-rich central cavity (central annulus). Thus, their amphiphilic properties qualify them to encapsulate the water-insoluble harmala alkaloids, within their central cavities.21,41 The ability to encapsulate high concentrations of drugs inside nanovesicles supports sustained drug release.42
2.2.2. Morphological Features
The TEM images presented in Figure 4 show HARF loaded in amphiphilic p-SC6 nanocapsules, which have spherical shapes with smooth surfaces.
Figure 4.
TEM image for H/p-SC6 nanocapsules at a scale of 200 nm showing sizes of (A) 275.43 nm and (B) 250.41 nm.
The self-assembly of amphiphilic p-SC6 is capable of producing spherical nanovesicles in aqueous solutions. The unique amphiphilic nature of p-SC6 permits the solubilization of the harmala alkaloid-rich fraction in the inner hydrophobic core while revealing its hydrophilic sulfonate groups found at the upper rims to the exterior medium.21
2.2.3. FT-IR Analysis
The FT-IR spectra of unencapsulated HARF, p-SC6, and the designed nanocapsules were compared to demonstrate the encapsulation of HARF into the cavity of the p-SC6 (Figure 5). Three major sharp bands, for pure HARF, are observed at 3444.02 cm–1 (−OH stretching vibrations), 1639.55 cm–1 (C=O asymmetry stretching vibration), and 1014.59 cm–1 (C–O stretching). These spectral bands agree with those reported previously.43 Upon encapsulation of HARF in p-SC6 nanocapsules, it is observed that the C–O stretching band at 1014.59 cm–1 had disappeared compared to HARF. Thus, this could indicate the encapsulation of HARF inside the cavity of p-SC6.27
Figure 5.
Fourier transform infrared (FTIR) spectra of (A) pure HARF, (B) p-SC6, and (C) H/p-SC6 nanocapsules.
2.2.4. In Vitro Release Study
The release of harmala major alkaloids from p-SC6-based nanocapsules was studied at pH 7.4 of healthy cells and pH 5.5 of cancer cells (Figure 6A–C).44 The released three alkaloids (peganine, harmol, and harmine) were determined using the above-mentioned HPLC–DAD method. As expected from ζ-potential measurements, H/p-SC6 nanocapsules exhibited outstanding stability at pH 7.4 with about 28.3, 39.9, and 35.2% of the loaded harmala alkaloids released after 48 h at 37 °C. At pH 5.5, 89.2, 88.9, and 90.8% of the loaded harmala alkaloids were released after 48 h at 37 °C. These results support the ability of self-assembled calixarenes to release their cargo selectively by a pH-triggered mechanism at a typical cancerous tissue pH of 5.5.45
Figure 6.

Time-dependent release profiles of the three harmala alkaloids from H/p-SC6 nanocapsules: (A) peganine, (B) harmol, and (C) harmine at 37 °C, into pH 5.5 (triangle) and pH 7.4 (square) phosphate buffer media.
These designed nanocapsules hold much promise for future cancer therapy because of their capability to release the encapsulated drugs inside the tumor tissue while shielding them from early decomposition in the systemic circulation of pH 7.4.
2.2.5. Antioxidant Activity
The antioxidant activity of free HARF and H/p-SC6 nanocapsules was investigated utilizing 2,2-diphenyl-1-picrylhydrazyl radical (DPPH) free radical scavenging assay. Trolox, a powerful antioxidant, was used as a positive control and showed an IC50 of 56.82 ± 0.87 μg/mL (Figure 7). It was shown that the IC50 of the H/p-SC6 nanocapsules (30.10 ± 1.30 μg/mL) exhibited about 5 times that of the free HARF (169.3 ± 7.2 μg/mL), as presented in Figure 7.
Figure 7.

IC50 values for DPPH radical scavenging assay of Trolox (positive control), harmala alkaloid-rich fraction (HARF), and H/p-SC6 nanocapsules.
The nanocapsules’ enhanced antioxidant activity compared to harmala alkaloid-rich fraction might be attributed to enhancing the hydrophilic nature of harmala alkaloid-rich fraction upon its encapsulation within the p-SC6 self-assemblies.46,47 Additionally, calixarenes are reported to have antioxidant properties combined with the antioxidant abilities of harmala alkaloid-rich fraction, giving rise to more pronounced free radical scavenging activities.48 These findings are very promising because powerful antioxidants have many biomedical applications and might play a vital role in protecting normal cells, especially during cancer treatment.
2.2.6. In Vitro Cell Viability Assay
The cytotoxicity of p-SC6, free HARF, and H/p-SC6 nanocapsules was assessed by SRB assay and ovarian cancer cells (SKOV-3), breast adenocarcinoma cells (MCF-7), and normal human skin fibroblasts.23,24p-SC6 was used as host control, and it showed a negligible decline in cell viability, and HARF was used as the positive control. After 48 h incubation, the harmala extract nanocapsules (H/p-SC6) exerted remarkable in vitro cytotoxicity compared to the free powder extract (positive control). The cytotoxic activities (IC50 in μg/mL, computed by Sigma plot) of p-SC6, free HARF, and H/p-SC6 nanocapsules against cancer noncancer cell lines are presented in Table 3. The IC50 values of H/p-SC6 were about 1.2- and 3-fold less than that of the HARF against SKOV-3 and MCF-7 cells, respectively. On the other hand, testing H/p-SC6 nanocapsules on HSF noncancer cells confirmed their biocompatibility compared to free harmala seed fraction. The previous studies reported the ability of different host molecules to encapsulate anticancer drugs and selectively deliver them to cancer cells with minimal toxic effects on normal cells.23,24,45−47,49−51 These results agree with the in vitro release study demonstrating the pH-triggered release of the HARF from the self-assembled H/p-SC6 nanocapsules. Cancerous cells have an acidic pH of 5.7, normal cells possess a pH of 7.4, whereas late endosomes and lysosomes have pH values of 4.5–5.5. This pH variation is vital because the pH-sensitive carriers would selectively release their cargo in the acidic tumor microenvironment.45,52,53 Additionally, HARF showed increased cytotoxic activities as its encapsulation within the p-SC6 nanocapsules overcame the extract’s hydrophobic nature and improved its bioavailability.23,24,46 This study also highlights the importance of using P. harmala seed alkaloid-rich fraction as a possible natural anticancer drug. Harmala alkaloid-rich fraction’s anticancer activity is attributed to the β-carboline alkaloids that inhibit DNA topoisomerases via DNA intercalation and induction of apoptotic pathways.54,55 Additionally, harmine alkaloid has been shown to possess antiangiogenic activity, reduce the expression of numerous proangiogenic factors, and diminish the propagation of vascular endothelial cells.56 Thus, these findings shed more light on the significance of using self-assembled p-SC6 as a possible supramolecular carrier for the natural promising anticancer agent, P. harmala seed alkaloid-rich fraction.
Table 3. In Vitro Cytotoxic Activities of p-SC6, Harmala Alkaloid-Rich Fraction, and H/p-SC6 Nanocapsules against SKOV-3, MCF-7, and Human Skin Fibroblastsa.
|
in
vitro cytotoxic activity (IC50 in μg/mL) |
|||
|---|---|---|---|
| cells | p-SC6 | harmala alkaloid-rich fraction (positive control) | H/p-SC6 |
| SKOV-3 | >300 | 6.7 ± 1.6 | 5 ± 1.3 |
| MCF-7 | >220 | 6.7 ± 0.7 | 2.3 ± 1.1 |
| human skin fibroblasts | >300 | 4.6 ± 1.5 | >300 |
The treatment period was 48 h. Data represent the mean ± standard deviation of triplicate.
3. Conclusions
In the present work, the harmala alkaloid-rich fraction was extracted from P. harmala seeds, and 15 alkaloids were annotated using ultraperformance liquid chromatography–electrospray ionization–tandem mass spectrometry (UPLC/ESI-MS). Reversed-phase high-performance liquid chromatography coupled with diode array detector (HPLC–DAD) analysis of the alkaloid-rich fraction revealed three major peaks: peganine, harmol, and harmine. This fraction was encapsulated in amphiphilic p-SC6 self-assembled nanovesicles employing a thin-film hydration approach. The designed nanocapsules possessed an average particle size of 264.77 ± 10.56 nm, a surface charge of −30.33 ± 2.15 mV, and were found to encapsulate 89.34 ± 1.41, 74.39 ± 1.3, and 76.1 ± 1.66% of the three harmala alkaloids, peganine, harmol, and harmine, respectively. In vitro drug release experiments showed the potential of the designed nanocapsules to release their cargo in mildly acidic media. This is very promising for future cancer therapy because of their ability to release the encapsulated drugs inside the acidic cancer tissue while keeping them stable in the systemic circulation of pH 7.4. The antioxidant activity of the H/p-SC6 nanocapsules was 5 times that of harmala alkaloid-rich fraction. Additionally, the cytotoxicity study revealed that the IC50 values of the designed nanocapsules were about 1.2- and 3-fold less than that demonstrated by harmala alkaloid-rich fraction against SKOV-3 and MCF-7 cells, respectively. The prepared nanocapsules were found to be biocompatible when tested on human skin fibroblasts. Thus, encapsulation of harmala alkaloid-rich fraction into self-assembled supramolecular nanocapsules dramatically enhances the natural anticancer and antioxidant effects of harmala alkaloids.
4. Materials and Methods
4.1. Materials
p-Sulfocalix[6]arene was purchased from WuXi LabNetwork, China. Diphenyl-1-picrylhydrazyl radical (DPPH), Harmine standard, HPLC-grade deionized water, chemicals, and solvents were obtained from Sigma-Aldrich (St. Louis, MO). Streptomycin, penicillin, fetal bovine serum, trichloroacetic acid (TCA), Dulbecco’s modified Eagle’s medium (DMEM) SRB, and tris(hydroxymethyl)aminomethane (TRIS) were obtained from Lonza, Basel, Switzerland.
4.2. Plant Material
Dried mature seeds of P. harmala L. were purchased from the Egyptian local market. A voucher specimen was deposited (18.1.17) at the Department of Pharmacognosy Herbarium, Faculty of Pharmacy, Cairo University (Egypt).
4.3. Methods
4.3.1. Extraction and Isolation of Major P. harmala Alkaloids
P. harmala seeds (2.5 kg) were ground to a fine powder and then extracted by soaking in 70% ethanol (3 × 5 L) at ambient temperature overnight. The ethanol extracts were filtered, combined, and evaporated under reduced pressure to yield a dark reddish-brown viscous residue (520 g). The concentrated extract was then dissolved in 5% HCl (2 L), filtered, and partitioned with dichloromethane (4 × 300 mL). The aqueous acid layer’s pH was adjusted to pH 9 using NH4OH and then extracted with dichloromethane (4 × 500 mL). The dichloromethane layer was washed with water and then evaporated under reduced pressure at 40 °C to yield 60 g of reddish-brown powder (harmala alkaloid-rich fraction).16
4.3.2. HPLC–DAD Analysis of the Three Major Harmala Alkaloids
The analysis of P. harmala alkaloids was performed, according to our previously reported method,16 by RP-HPLC using a Waters LC 2695 coupled to a Waters 996 diode array detector (DAD). A 100 × 4.6 mm2 id, 5 μm, XTerra MS-C18 column (Agilent Technologies) was used for analysis. Chromatographic conditions were water supplemented with triethylamine (pH 8.6) (A) and acetonitrile (B). The gradient was programmed from 0% B (100% A) to 20% B in 4 min, then 30% B at 9 min, 50% B at 14 min, and finally to 60% B at 16 min. The separation was followed by a 2 min washing procedure with 100% B and a reequilibration period of 4 min. Flow rate: 1 mL/min, column temperature: 25 °C, injection volume: 20 μL, and absorbance detection: 320 nm. Under these conditions, the elution order was peganine, harmol, and harmaline. These peaks were compared to standard harmine, and the isolated alkaloids were peganine, harmol, and harmine when injected individually on HPLC (Figure 1). The isolated ones were annotated by their mass and UV spectra (Figures S1 and S2). Calibration curves were constructed for peganine, harmol, and harmine to be used in the measurement of encapsulation efficiency and in vitro release studies.
4.3.3. UPLC-MS/MS Analysis of Harmala Alkaloids
The harmala alkaloid-rich fraction was analyzed using UPLC/ESI-MS. The sample was dissolved in methanol for HPLC at a final concentration of 100 μg/mL. Then, it was filtered using a membrane disk filter (0.2 μm) before subjecting to LC-ESI-MS analysis on UPLC/ESI-MS, ACQUITY UPLC System (Waters Corporation) using ACQUITY UPLC-BEH C18 (1.7 μm, 2.1 × 50 mm2) column and injection volume of 10 μL. The solvent system consisted of (A) water containing 0.1% formic acid and (B) methanol containing 0.1% formic acid. Elution was done using gradient mobile phase starting from 90% A: 10% B that was maintained for 2 min, it reached 70% A at 5 min, then 30% A at 15 min, 10% A at 22 min which was maintained for 3 min and then was changed to reach 100% B at 26 min and was upheld for 3 min and then back to the initial composition at 32 min. The flow rate was 0.2 mL/min. The parameters for analysis were carried out in positive-ion acquisition mode using a XEVO TQD triple quadrupole mass spectrometer (Waters Corporation, Milford, MA). The source temperature, cone voltage, capillary voltage, and desolvation temperature were 150 °C, 30 eV, 3 kV, and 440 °C, respectively. The cone gas flow rate was 50 L/h, and the desolvation gas flow rate was 900 L/h. Mass spectra were detected in the ESI between m/z 100 and 1000. The peaks and spectra were processed using the Maslynx 4.1 software and tentatively identified by comparing their retention times (Rt) and mass spectra with reported data.16
4.3.4. Preparation of Harmala Alkaloid-Rich Fraction-Loaded p-Sulfonatocalix[6]arene Nanocapsules (H/p-SC6)
H/p-SC6 nanocapsules were produced using the thin-film hydration approach as described elsewhere27,49,50 with some modifications. Briefly, p-SC6 (57 mg) was mixed with HARF (1 mg) in a sufficient volume of methanol in a round-bottom flask. The round-bottom flask was exposed to evaporation under vacuum for 60 min at 40 °C, utilizing a Laborotay 4000 rotary evaporator (Heidolph Instruments, Schwabach, Germany) equipped with a vacuum pump (KNF Neuberger GmbH, Freiburg, Germany), leaving a thin film. Then, hydration was conducted using phosphate-buffered saline (PBS, pH 7.4) in a rotary evaporator without using vacuum for 30 min at 40 °C. Finally, sonication for 1 min of the produced suspension was carried out employing a bath sonicator (Elmasonic P30 H, Elma Hans Schmidbauer, Singen, Germany).
4.3.5. Characterization of the Designed H/p-SC6 Nanocapsules
The average particle size and polydispersity index (PDI) of the prepared nanocapsules were determined using dynamic light scattering employing Zetasizer Nano ZS (Malvern Instruments Herrenberg, Germany).57 The instrument is equipped with a 10 mW HeNe laser allowing for the measurements to be performed at a wavelength of 633 nm and a detection angle of 173° backscatter. The ζ-potential of the nanocapsules was determined via laser Doppler velocimetry in a clear disposable folded capillary cell (DTS1070, Malvern Instruments).
The morphological features of the nanocapsules were studied using transmission electron microscopy (TEM), employing a JEOL-JEM 2100 electron microscope operating at 160 kV. A 50 μL aliquot of the nanovesicles diluted to 1:2 (v/v) with PBS were stained with 2% aqueous phosphotungstic acid. This mixture was placed and dried over a carbon-coated copper 200 mesh grid, imagined, and photographed.
The FT-IR spectra of unencapsulated HARF, p-SC6, and the designed nanocapsules were subjected to Fourier transform infrared (FTIR) spectroscopy employing FTIR-8400s (Shimadzu, Japan). Samples were first compressed with KBr into disks, scanned, and spectra were recorded in the range of 500–4000 cm–1.
4.3.6. Encapsulation Efficiency
The encapsulation efficiency (EE) of H/p-SC6 nanocapsules was determined by centrifugation at 14 000 rpm for 90 min at 4 °C, followed by ultrafiltration to eliminate the harmala alkaloids loaded pellet. The quantity of the harmala alkaloids unencapsulated in the ultrafiltrate was detected using the HPLC protocol described earlier. The EE of the H/p-SC6 nanocapsules was determined using eq 1(27,58)
| 1 |
4.3.7. In Vitro Release Study under Different pH Conditions
The release rate of harmala alkaloids from H/p-SC6 nanocapsules was studied using the dialysis membrane method at pH 5.5 and 7.4. Briefly, 1 mL of the sample was loaded to a dialysis bag (cutoff molecular weight, 3500 Da). The dialysis bag was inserted into 50 mL of PBS at pH 7.4 or 5.5, with 1% Tween and 0.5% FBS in a proper jar. The whole system was fixed in a shaking incubator (Jeio Tech SI-300, Seoul, Korea), rotating at 100 rpm with temperature adjusted to 37 °C. At specific time intervals, a 1 mL aliquot of the sample was withdrawn for analysis by the above-described HPLC method and immediately replaced with another equal volume of warmed buffer.
4.3.8. Determination of Total Antioxidant Activity
The antioxidant activity of HARF and H/p-SC6 nanocapsules was evaluated using DPPH (2,2-diphenyl-1-picryl-hydrazyl-hydrate) free radical scavenging assay as previously reported by Boly et al.59 with minor modification. Briefly, 100 μL of freshly prepared DPPH reagent (0.1% in methanol) was added to 100 μL of the sample in a 96-well plate (n = 6) and the reaction was left at room temperature for 30 min in the dark. Afterward, the color intensity of the DPPH resulting from its reduction was recorded at 540 nm using a microplate reader FluoStar Omega (Ortenberg, Germany). Data are represented as mean ± SD according to eq 2
| 2 |
where Acontrol is the absorbance of the DPPH and Asample is the absorbance of the sample.
Trolox was considered a reference standard, and DPPH solution was used as the control. Data were analyzed using Microsoft Excel, and the IC50 value was calculated using GraphPad Prism 5 by converting the concentrations to their logarithmic value and selecting nonlinear inhibitor regression equation [log (inhibitor) versus normalized response – variable slope equation].60
4.3.9. Cell Viability Assay
4.3.9.1. Cell Culture
Ovarian cancer cells (SKOV-3), breast adenocarcinoma cells (MCF-7), and human skin fibroblasts healthy cells were obtained from American Type Culture Collection, (University Boulevard, Manassas, VA 20110) and maintained in DMEM supplemented with streptomycin (100 mg/mL), penicillin (100 units/mL), and 10% heat-inactivated fetal bovine serum. Cells were incubated in humidified 5% (v/v) CO2 at 37 °C.
4.3.9.2. Sulforhodamine B Colorimetric Assay
Ovarian cancer cells (SKOV-3), breast adenocarcinoma cells (MCF-7), and human skin fibroblasts were treated with different concentrations of p-SC6, free HARF (positive control), and H/p-SC6. The cell viability of either cancerous or noncancerous cells was tested using the SRB assay, and the IC50 (in μg/mL) value was computed using our methods described previously.23,24,61,62
All trials were conducted in triplicates, and data are demonstrated as mean ± standard deviation.
Acknowledgments
The authors thank the Chemistry Department at the American University in Cairo for the provision of resources for the project.
Supporting Information Available
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsomega.1c00455.
ESI/MS spectra in positive-ion mode of peganine, harmol, and harmine (Figure S1); UV spectra of peganine, harmol, and harmine (Figure S2); and MS/MS fragmentation pattern of selected alkaloids; vasicine, harmalol, hydroxylated harmine, harmaline, harmine, and harmol, detected in harmala alkaloid-rich fraction by ultraperformance liquid chromatography–electrospray ionization–tandem mass spectrometry (UPLC/ESI-MS) using positive-ion mode (Figure S3) (PDF)
Author Contributions
∥ S.A.F. and M.Y.I. contributed equally to this work.
This project was funded by a grant from the American University in Cairo to H.M.E.-S.A
The authors declare no competing financial interest.
Supplementary Material
References
- Ferlay J.; Soerjomataram I.; Dikshit R.; Eser S.; Mathers C.; Rebelo M.; Parkin D. M.; Forman D.; Bray F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in Globocan 2012. Int. J. Cancer 2015, 136, E359–E386. 10.1002/ijc.29210. [DOI] [PubMed] [Google Scholar]
- Priestman T.Cancer Chemotherapy in Clinical Practice, 2nd ed.; Springer-Verlag: London, 2012. [Google Scholar]
- Sriram D.; Yogeeswari P.. Medicinal Chemistry, 2nd ed.; Dorling Kindersley: New Delhi, 2010. [Google Scholar]
- Jakupec M. A.; Galanski M.; Keppler B. K. Tumor-inhibiting platinum complexes-state of the art and future perspectives. Rev. Physiol., Biochem. Pharmacol. 2003, 146, 1–53. 10.1007/s10254-002-0001-x. [DOI] [PubMed] [Google Scholar]
- Kanzawa F.; Matsushima Y.; Nakano H.; Nakagawa K.; Takahashi H.; Sasaki Y.; Saijo N. Antitumor activity of a new platinum compound (glycolate o,o’) diammineplatinum (II) (254-S), against non-small cell lung carcinoma grown in a human tumor clonogenic assay system. Anticancer Res. 1988, 8, 323–327. [PubMed] [Google Scholar]
- Ostos F.; Lebron J.; Moya M.; Lopez-Lopez M.; Sanchez A.; Clavero A.; Garcia-Calderon C.; Rosado I.; Lopez-Cornejo P. P-Sulfocalix[6]arene as Nanocarrier for Controlled Delivery of Doxorubicin. Chem. - Asian J. 2017, 12, 679–689. 10.1002/asia.201601713. [DOI] [PubMed] [Google Scholar]
- Mann K.; Kullberg M. Trastuzumab-targeted gene delivery to Her2-overexpressing breast cancer cells. Cancer Gene Ther. 2016, 23, 221–228. 10.1038/cgt.2016.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J.; Lan C. Q.; Post M.; Simard B.; Deslandes Y.; Hsieh T. H. Design of nanoparticles as drug carriers for cancer therapy. Cancer Genomics Proteomics 2006, 3, 147–157. [PubMed] [Google Scholar]
- Kwon G. S. Polymeric micelles for delivery of poorly water soluble compounds. Crit. Rev. Ther. Drug Carrier Syst. 2003, 20, 357–403. 10.1615/CritRevTherDrugCarrierSyst.v20.i5.20. [DOI] [PubMed] [Google Scholar]
- Luo Y.; Prestwich G. D. Cancer-targeted polymeric drugs. Curr. Cancer Drug Targets 2002, 2, 209–226. 10.2174/1568009023333836. [DOI] [PubMed] [Google Scholar]
- Mukherjee A. K.; Basu S.; Sarkar N.; Ghosh A. C. Advances in cancer therapy with plant based natural products. Curr. Med. Chem. 2001, 8, 1467–1486. 10.2174/0929867013372094. [DOI] [PubMed] [Google Scholar]
- Smyth M. J.; Hayakawa Y.; Takeda K.; Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer. Nat. Rev. Cancer 2002, 2, 850–861. 10.1038/nrc928. [DOI] [PubMed] [Google Scholar]
- Newman D. J.; Cragg G. M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. 10.1021/acs.jnatprod.9b01285. [DOI] [PubMed] [Google Scholar]
- Fahmy S. A.; Preiss E.; Bakowsky U.; Azzazy H. M. E.-S.. Palladium Nanoparticles Fabricated by Green Chemistry: Promising Chemotherapeutic, Antioxidant and Antimicrobial Agents. Materials 2020133661. 10.3390/ma13173661 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fahmy S. A.; Mamdouh W. Garlic oil–loaded PLGA nanoparticles with controllable size and shape and enhanced antibacterial activities. J. Appl. Polym. Sci. 2018, 135, 46133 10.1002/app.46133. [DOI] [Google Scholar]
- Shaheen H. A.; Issa M. Y. In vitro and in vivo activity of Peganum harmala L. alkaloids against phytopathogenic bacteria. Sci. Hortic. 2020, 264, 108940 10.1016/j.scienta.2019.108940. [DOI] [Google Scholar]
- Cao R.; Peng W.; Wang Z.; Xu A. β-Carboline alkaloids: biochemical and pharmacological functions. Curr. Med. Chem. 2007, 14, 479–500. 10.2174/092986707779940998. [DOI] [PubMed] [Google Scholar]
- Herraiz T.; González D.; Ancín-Azpilicueta C.; Arán V.; Guillén H. β-Carboline alkaloids in Peganum harmala and inhibition of human monoamine oxidase (MAO). Food Chem. Toxicol. 2010, 48, 839–845. 10.1016/j.fct.2009.12.019. [DOI] [PubMed] [Google Scholar]
- Wink M.; Schmeller T.; Latz-Brüning B. Modes of action of allelochemical alkaloids: interaction with neuroreceptors, DNA, and other molecular targets. J. Chem. Ecol. 1998, 24, 1881–1937. 10.1023/A:1022315802264. [DOI] [Google Scholar]
- El-Shafie S.; Fahmy S. A.; Ziko L.; Elzahed N.; Shoeib T.; Kakarougkas A. Encapsulation of Nedaplatin in Novel PEGylated Liposomes Increases Its Cytotoxicity and Genotoxicity against A549 and U2OS Human Cancer Cells. Pharmaceutics 2020, 12, 863 10.3390/pharmaceutics12090863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fahmy S. A.; Brüßler J.; Alawak M.; El-Sayed M. M. H.; Bakowsky U.; Shoeib T. Chemotherapy Based on Supramolecular Chemistry: A Promising Strategy in Cancer Therapy. Pharmaceutics 2019, 11, 292 10.3390/pharmaceutics11060292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fahmy S. A.; Ponte F.; Abd El-Rahman M. K.; Russo N.; Sicilia E.; Shoeib T. Investigation of the host-guest complexation between 4-sulfocalix[4]arene and nedaplatin for potential use in drug delivery. Spectrochim. Acta, Part A 2018, 193, 528–536. 10.1016/j.saa.2017.12.070. [DOI] [PubMed] [Google Scholar]
- Fahmy S. A.; Ponte F.; Sicilia E.; Azzazy H. M. E.-S. Experimental and Computational Investigations of Carboplatin Supramolecular Complexes. ACS Omega 2020, 5, 31456–31466. 10.1021/acsomega.0c05168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fahmy S. A.; Ponte F.; Fawzy I. M.; Sicilia E.; Bakowsky U.; Azzazy H.-S. Host-Guest Complexation of Oxaliplatin and Para-Sulfonatocalix[n]Arenes for Potential Use in Cancer Therapy. Molecules 2020, 25, 5926 10.3390/molecules25245926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravoo B. J.Vesicles in Supermolecular Chemistry, Supermolecular Chemistry: From Molecules to Nanomaterials; Gale P. A.; Steed J. W., Eds.; John Wiley & Sons: New York, NY, 2012; pp 354–379. [Google Scholar]
- Markowitz M. A.; Bielski R.; Regen S. L. Ultrathin monolayers and vesicular membranes from calix[6] arenes. Langmuir 1989, 5, 276–278. 10.1021/la00085a054. [DOI] [Google Scholar]
- Chen M.-X.; Li T.; Peng S.; Tao D. Supramolecular nanocapsules from the self-assembly of amphiphilic calixarene as a carrier for paclitaxel. New J. Chem. 2016, 40, 9923–9929. 10.1039/C6NJ01986B. [DOI] [Google Scholar]
- Herraiz T.; Guillén H.; Arán V. J.; Salgado A. Identification, occurrence and activity of quinazoline alkaloids in Peganum harmala. Food Chem. Toxicol. 2017, 103, 261–269. 10.1016/j.fct.2017.03.010. [DOI] [PubMed] [Google Scholar]
- Liu L.; Zhao T.; Cheng X. M.; Wang C. H.; Wang Z. T. Characterization and determination of trace alkaloids in seeds extracts from Peganum harmala linn. Using LC-ESI-MS and HPLC. Acta Chromatogr. 2013, 25, 221–240. 10.1556/AChrom.25.2013.2.2. [DOI] [Google Scholar]
- Wang Z.; Kang D.; Jia X.; Zhang H.; Guo J.; Liu C.; Meng Q.; Liu W. Analysis of alkaloids from Peganum harmala L. sequential extracts by liquid chromatography coupled to ion mobility spectrometry. J. Chromatogr. B 2018, 1096, 73–79. 10.1016/j.jchromb.2018.08.021. [DOI] [PubMed] [Google Scholar]
- Telezhenetskaya M. V.; Khashimov K. N.; Yunusov S. Y. Peganol—A new alkaloid from Peganum harmala. Chem. Nat. Compd. 1971, 7, 835 10.1007/BF00567971. [DOI] [Google Scholar]
- Wang C. H.; Zeng H.; Wang Y. H.; Li C.; Cheng J.; Ye Z. J.; He X. J. Antitumor quinazoline alkaloids from the seeds of Peganum harmala. J. Asian Nat. Prod. Res. 2015, 17, 595–600. 10.1080/10286020.2015.1042373. [DOI] [PubMed] [Google Scholar]
- Khashimov K. N.; Telezhenetskaya M. V.; Rashkes Y. V.; Yunusov S. Y. Pegamine: A new alkaloid from Peganum harmala. Chem. Nat. Compd. 1970, 6, 462–464. 10.1007/BF00564251. [DOI] [Google Scholar]
- Zhao T.; Zheng S. S.; Zhang B. F.; Li Y. Y.; Bligh S. A.; Wang C. H.; Wang Z. T. Metabolic pathways of the psychotropic-carboline alkaloids, harmaline and harmine, by liquid chromatography/mass spectrometry and NMR spectroscopy. Food Chem. 2012, 134, 1096–1105. 10.1016/j.foodchem.2012.03.024. [DOI] [PubMed] [Google Scholar]
- Zhang L.; Teng L.; Gong C.; Liu W.; Cheng X.; Gu S.; Deng Z.; Wang Z.; Wang C. Simultaneous determination of harmine, harmaline and their metabolites harmol and harmalol in beagle dog plasma by UPLC–ESI-MS/MS and its application to a pharmacokinetic study. J. Pharm. Biomed. Anal. 2013, 85, 162–168. 10.1016/j.jpba.2013.07.019. [DOI] [PubMed] [Google Scholar]
- METLIN Metabolite and Chemical Entity Database. https://metlin.scripps.edu/landing_page.php?pgcontent=mainPage. (accessed on April 1, 2021).
- Siddiqui S.; Khan O. Y.; Faizi S.; Siddiqui B. S. Studies in the chemical constituents of the seeds of Peganum harmala: isolation and structure elucidation of two β-carboline lactams―harmalanine and harmalacidine. Heterocycles 1988, 27, 1401–1410. 10.3987/COM-88-4490. [DOI] [Google Scholar]
- Rao K.; Roome T.; Aziz S.; Razzak A.; Abbas G.; Imran M.; Jabri T.; Gul J.; Hussain M.; Sikandar B.; et al. Bergenin loaded gum xanthan stabilized silver nanoparticles suppress synovial inflammation through modulation of the immune response and oxidative stress in adjuvant induced arthritic rats. J. Mater. Chem. B 2018, 6, 4486–4501. 10.1039/C8TB00672E. [DOI] [PubMed] [Google Scholar]
- Imran M.; Shah M. R.; Ullah F.; Ullah S.; Sadiq A.; Ali I.; Ahmed F.; Nawaz W. Double-tailed acyl glycoside niosomal nanocarrier for enhanced oral bioavailability of Cefixime. Artif. Cells, Nanomed., Biotechnol. 2017, 45, 1440–1451. 10.1080/21691401.2016.1246451. [DOI] [PubMed] [Google Scholar]
- Cho K.; Wang X.; Nie S.; Chen Z. G.; Shin D. M. Therapeutic Nanoparticles for Drug Delivery in Cancer. Clin. Cancer Res. 2008, 14, 1310–1316. 10.1158/1078-0432.CCR-07-1441. [DOI] [PubMed] [Google Scholar]
- Dong J.; Xun Z.; Zeng Y.; Yu T.; Han Y.; Chen J.; Li Y. Y.; Yang G.; Li Y. A Versatile and Robust Vesicle Based on a Photocleavable Surfactant for Two-Photon-Tuned Release. Chem. - Eur. J. 2013, 19, 7931–7936. 10.1002/chem.201300526. [DOI] [PubMed] [Google Scholar]
- Ullah S.; Shah M. R.; Shoaib M.; Imran M.; Elhissi A. M.; Ahmad F.; Ali I.; Shah S. W. A. Development of a biocompatible creatinine-based niosomal delivery system for enhanced oral bioavailability of clarithromycin. Drug Delivery 2016, 23, 3480–3491. 10.1080/10717544.2016.1196768. [DOI] [PubMed] [Google Scholar]
- Azizi M.; Sedaghat S.; Tahvildari K.; Derakhshi P.; Ghaemi A. Synthesis of silver nanoparticles using Peganum harmala extract as a green route. Green Chem. Lett. Rev. 2017, 10, 420–427. 10.1080/17518253.2017.1395081. [DOI] [Google Scholar]
- Tannock I. F.; Rotin D. Acid pH in tumors and its potential for therapeutic exploitation. Cancer Res. 1989, 49, 4373–4384. [PubMed] [Google Scholar]
- Hamid S. A.; Bunnori N. M.; Ishola N. M.; Ali Y. Applications of calixarenes in cancer chemotherapy: Facts and perspectives. Drug Des., Dev. Ther. 2015, 9, 2831–2838. 10.2147/DDDT.S83213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang D.; Zhang J.; Jiang K.; Li K.; Cong Y.; Pu S.; Jin Y.; Lin J. Preparation, characterisation and antitumour activity of β-, γ-and HP-β-cyclodextrin inclusion complexes of oxaliplatin. Spectrochim. Acta, Part A 2016, 152, 501–508. 10.1016/j.saa.2015.07.088. [DOI] [PubMed] [Google Scholar]
- Fahmy S. A.; Brüßler J.; Ponte F.; Abd El-Rahman M. K.; Russo N.; Sicilia E.; Bakowsky U.; Shoeib T. A study on the physicochemical properties and cytotoxic activity of p-sulfocalix[4]arene-nedaplatin complex. J. Phys.: Conf. Ser. 2019, 1310, 012011 10.1088/1742-6596/1310/1/012011. [DOI] [Google Scholar]
- Lee J.-S.; Song I.-H.; Shinde P. B.; Nimse S. B. Macrocycles and Supramolecules as Antioxidants: Excellent Scaffolds for Development of Potential Therapeutic Agents. Antioxidants 2020, 9, 859 10.3390/antiox9090859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M.; Mao L.; Chen M.; Li M.; Wang K.; Mo J. Characterization of an Amphiphilic Phosphonated Calixarene Carrier Loaded with Carboplatin and Paclitaxel: A Preliminary Study to Treat Colon Cancer in vitro and in vivo. Front. Bioeng. Biotechnol. 2019, 7, 238 10.3389/fbioe.2019.00238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mo J.; Wang L.; Huang X.; Lu B.; Zou C.; Wei L.; et al. Multifunctional nanoparticles for co-delivery of paclitaxel and carboplatin against ovarian cancer by inactivating the JMJD3-HER2 axis. Nanoscale 2017, 9, 13142–13152. 10.1039/C7NR04473A. [DOI] [PubMed] [Google Scholar]
- Liu R.; Lai Y.; He B.; Li Y.; Wang G.; Chang S.; Gu Z. Supramolecular nanoparticles generated by the self-assembly of polyrotaxanes for antitumor drug delivery. Int. J. Nanomed. 2012, 7, 5249–5258. 10.2147/IJN.S33649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bae Y. M.; Park Y. I.; Nam S. H.; Kim J. H.; Lee K.; Kim H. M.; et al. Endocytosis, intracellular transport, and exocytosis of lanthanide-doped upconverting nanoparticles in single living cells. Biomaterials 2012, 33, 9080–9086. 10.1016/j.biomaterials.2012.08.039. [DOI] [PubMed] [Google Scholar]
- Iversen T. G.; Skotland T.; Sandvig K. Endocytosis and intracellular transport of nanoparticles: present knowledge and need for future studies. Nano Today 2011, 6, 176–185. 10.1016/j.nantod.2011.02.003. [DOI] [Google Scholar]
- Park S. Y.; Kim Y. H.; Kim Y. H.; Park G.; Lee S. J. Beta-carboline alkaloids harmaline and harmalol induce melanogenesis through p38 mitogen-activated protein kinase in B16F10 mouse melanoma cells. BMB Rep. 2010, 43, 824–829. 10.5483/BMBRep.2010.43.12.824. [DOI] [PubMed] [Google Scholar]
- Cao M. R.; Li Q.; Liu Z. L.; Liu H. H.; Wang W.; Liao X. L.; Pan Y. L.; Jiang J. W. Harmine induces apoptosis in HepG2 cells via mitochondrial signaling pathway. Hepatobiliary Pancreatic Dis. Int. 2011, 10, 599–604. 10.1016/S1499-3872(11)60102-1. [DOI] [PubMed] [Google Scholar]
- Hashemi S.; Shabani S.; Tehrani S. S. H.; Rabiei Z.; Enferadi S. T.; Vannozzi G. P. Peganum harmala L.’s anti-growth effect on a breast cancer cell line. J. Appl. Biotechnol. Rep. 2015, 8, 138–143. 10.1016/j.btre.2015.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pinnapireddy S. R.; Duse L.; Strehlow B.; Schäfer J.; Bakowsky U. Composite liposome-PEI/nucleic acid lipopolyplexes for safe and efficient gene delivery and gene knockdown. Colloids Surf., B 2017, 158, 93–101. 10.1016/j.colsurfb.2017.06.022. [DOI] [PubMed] [Google Scholar]
- Qiu L.; Jing N.; Jin Y. Preparation and in vitro evaluation of liposomal chloroquine diphosphate loaded by a transmembrane pH-gradient method. Int. J. Pharm. 2008, 361, 56–63. 10.1016/j.ijpharm.2008.05.010. [DOI] [PubMed] [Google Scholar]
- Boly R.; Lamkami T.; Lompo M.; Dubois J.; Guissou I. DPPH Free Radical Scavenging Activity of Two Extracts from Agelanthus dodoneifolius (Loranthaceae) Leaves. Int. J. Toxicol. Pharmacol. Res. 2016, 8, 29–34. [Google Scholar]
- Chen Z.; Bertin R.; Froldi G. EC50 estimation of antioxidant activity in DPPH assay using several statistical programs. Food Chem. 2013, 138, 414–420. 10.1016/j.foodchem.2012.11.001. [DOI] [PubMed] [Google Scholar]
- Fahmy S. A.; Fawzy I. M.; Saleh B. M.; Issa M. Y.; Bakowsky U.; Azzazy H. M. E.-S. Green Synthesis of Platinum and Palladium Nanoparticles Using Peganum harmala L. Seed Alkaloids: Biological and Computational Studies. Nanomaterials 2021, 11, 965. 10.3390/nano11040965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vichai V.; Kirtikara K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 2006, 1, 1112–1116. 10.1038/nprot.2006.179. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




