Skip to main content
Journal of Food and Drug Analysis logoLink to Journal of Food and Drug Analysis
. 2023 Jun 15;31(2):194–211. doi: 10.38212/2224-6614.3454

The therapeutic potential of curcumin and its related substances in turmeric: From raw material selection to application strategies

Kai-Yu Hsu a, Chi-Tang Ho b, Min-Hsiung Pan a,c,d,*
PMCID: PMC10281727  PMID: 37335161

Abstract

Turmeric (Curcuma longa L.) is a medicinal plant used extensively in Chinese and Indian traditional medicine as a home remedy for various diseases. It has been used for medical purposes for centuries. Today, turmeric has become one of the most popular medicinal herbs, spices, and functional supplements worldwide. Curcuminoids are linear diary-lheptanoids from the rhizomes that include curcumin and two related compounds: demethoxycurcumin and bisdemethoxycurcumin, which are the active components of the C. longa plant, play a crucial role in numerous functions. This review summarises the composition of turmeric and the properties of curcumin regarding its antioxidant, anti-inflammatory, anti-diabetic, anti-colorectal cancer, and other physiological activity. In addition, the dilemma of the application of curcumin due to its low water solubility and bioavailability was discussed. Finally, this article provides three novel application strategies based on previous studies: using curcumin analogues and related substances, gut microbiota regulation, and using curcumin-loaded exosome vesicles and turmeric-derived exosome-like vesicles to overcome application limitations.

Keywords: Anti-colorectal cancer, Anti-inflammation, Curcumin, Exosome vesicles, Gut microbiota

1. Introduction

Turmeric (Curcuma longa L.) is a medicinal plant that is used extensively in Chinese and Indian traditional medicine as a home remedy for various diseases. It is botanically related to ginger (Zingiberaceae family). It is a perennial plant with a short stem, large leaves, and rhizomes of various shapes that are often branched and have a brownish-yellow peel [1]. Today, turmeric is grown in several parts of the world, including Southeast Asia, China, and South America [2]. It has been used for medical purposes in India and China for many centuries to treat liver ailments [3] and other inflammation-related symptoms [4]. Turmeric has become one of the most popular medicinal herbs, spices, and functional supplements. The popularity of turmeric can be attributed to its pharmacological activity, including it antioxidant [5], antibacterial [6], anti-inflammatory [7], anti-tumour [8], and antiaging properties [9]. These functional activities of turmeric have been attributed to its rich curcuminoid content [1,2]. Curcuminoids (Fig. 1) are linear diary-lheptanoids from the rhizomes that include curcumin (CUR) and two related compounds: demethoxycurcumin (DMC) and bisdemethoxycurcumin (BDMC) [2]. Curcumin is a crystalline compound with a bright orange-yellow colour. It is often used as a food colouring and additive [2]. In the current international regulations, curcumin is considered a safe food additive. The World Health Organization (WHO) evaluated the acceptable daily intake (ADI) of curcumin as a food colouring additive in the range of 0–3 mg/kg [10]. The United States Food and Drug Administration (USFDA) has declared curcumin as ‘generally regarded as safe’ (GRAS). It is worth noting that most researchers consider curcumin to exhibit a wide variety of pharmacological properties and is relatively safe in animals and humans [11]; however, high doses (>50,000 ppm) ingested in order to overcome the low bioavailability of curcumin have some side effects and safety concerns [12]. Overall, if the bioavailability of curcumin can be effectively improved, it has the potential to improve or even cure diseases [13].

Fig. 1.

Fig. 1

Curcuminoids found in Curcuma longa, curcumin metabolites (reduction), and its degradation products (cleavage).

2. Composition of turmeric

Curcumin (C21H20O5) is also known as diferuloylmethane or (1E,6E )-1,7-bis(4-hydroxy-3-methoxyphenyl)hepta-1,6-diene-3,5-dione, DMC (C20H18O5) is also known as (1E,6E )-1-(4-hydroxy-3-methoxyphenyl)-7-(4-hydroxyphenyl)hepta-1,6-diene-3,5-dione, and BDMC (C19H16O4) is also known as (1E,6E )-1,7-bis(4-hydroxyphenyl)hepta-1,6-diene-3,5-dione. These three major curcuminoids are also found in other Curcuma species at different concentrations and proportions. Curcuminoids were only detected in a few species, such as C. longa and C. phaeocaulis, and geographical differences affect its content. In China, C. longa (Jianghuang) rhizomes from Pengzhou Sichuan contain the highest amount of curcuminoids (40.36 mg/g), which are almost 20-fold higher than those found in C. longa (Huangsi Yujin) collected from the same area [14]. In another study, C. longa rhizomes from the Zhengzhou pharmacies market exhibited a higher amount of curcuminoids (172.6 mg/g) [15]. Of note, curcuminoid content is related to geographical location; in Nepal, turmeric that is cultivated in warmer climates (Southern Nepal) has higher curcuminoid content than turmeric samples from cooler climates (Northern Nepal) [16]. In India, samples collected from different geographical regions also have different curcuminoid content; the main reason is the different environmental conditions across the Indian subcontinent. The maximum and minimum amount of curcuminoids were found to be present in samples of turmeric from Erode (South province, 50.27 mg/g) and Surat (West province, 14.08 mg/g) [17]. In Brazil, different samples in the same area contained different amounts of curcuminoids in the rhizome, from 18.2 mg/g to 23.3 mg/g. The curcumin content in the two samples is not much different and is mainly due to the difference in the content of DMC and BDMC [18]. Different Curcuma species may have different curcuminoid content, curcuminoids, and xanthorrhizol (XNT) content, which can be used as a specific marker to differentiate Curcuma xanthorriza and C. longa [19]. The guidelines of the Taiwan Herbal Pharmacopeia [20] and The Pharmacopoeia of the People's Republic of China 2020 Edition [21], two official compendiums of Chinese drugs, state that medicinal turmeric must contain more than 1% curcumin, and almost all turmeric rhizome samples meet this specification. However, this guideline is limited to the rhizome of turmeric. The curcumin content of the root is much lower than that of the rhizome [14]; therefore, the plant localisation or extract of medicinal turmeric must be considered for a standardised dose of curcumin [22]. Furthermore, ingested turmeric products may contain metabolites and degradation products of curcumin. Curcumin will easily degrade in neutral to alkaline solutions and is cleaved into ferulic acid, feruloyl methane, and vanillin (Fig. 1) [23]. Tetrahydrocurcumin (THC), hexahydrocurcumin (HHC), and octahydrocurcumin (OHC) (Fig. 1) are hydrogenated derivatives of curcumin [24] that appear as metabolites after curcumin is metabolised by cells or animals. Because of their structure, they are functionally closely related to curcumin [25]. In addition to the extraction and isolation of curcuminoids from turmeric, chemical synthesis is a strategy for obtaining curcuminoids [26,27]. The earliest record of chemical synthesis of curcumin was in 1913 [28]; since then, many studies have explored preparation methods of curcumin by chemical synthesis [26,29,30]. Unlike curcumin extracted from turmeric, which often contains a variety of curcuminoids, chemical synthesis can prepare specific, high-purity curcuminoids by adjusting conditions and precursors. This makes it possible to examine the chemical structure and mechanism of action of curcuminoids more precisely when discussing the function of curcuminoids.

3. Function of curcumin and curcuminoids

3.1. Antioxidant

As cells grow, oxygen consumption leads to the production of reactive oxygen species (ROS) [31]. ROS are forms of activated oxygen that include free radicals, such as superoxide anion radicals ( O2·-), hydroxyl radicals (OH), and non-free-radical species, such as H2O2 and singlet oxygen (1O2) [32]. ROS in cells cause the peroxidation of membrane lipids, leading to lipid peroxides. Furthermore, ROS can damage intracellular chemical composition, such as nucleic acids, lipids, proteins, and carbohydrates [25], which affect the inner workings of the cell. Generally speaking, antioxidant systems (e.g., the glutathione cycle and superoxide dismutase) in cells scavenge ROS and free radicals [33]; however, as the oxidative pressure increases, these systems may be overloaded and become ineffective. It has been confirmed that ROS production is directly related to many diseases [34]. Dietary antioxidants can protect the human body from free radicals and the effects of ROS. The curcuminoid structure contains several functional groups, including the β-diketone group, carbon–carbon double bonds, and phenyl rings containing hydroxyl and methoxy substituents [35]. Interestingly, the demethoxy and hydrogenated derivatives of curcumin, such as THC, HHC, and OHC, were remarkably more potent than curcumin in in vitro antioxidant assays [24]. Due to the loss of the ortho-methoxyphenolic group, BDMC does not have hydrogen donating activity [36].

3.2. Anti-inflammatory

Inducible nitric oxide synthase (iNOS) is one of the critical enzymes producing nitric oxide (NO) from the amino acid L-arginine, and iNOS-derived NO plays a crucial role in blood pressure regulation, inflammation, infection, and the progression of malignant diseases [37]. In macrophages, iNOS serves as a mediator of non-specific host defence and plays an essential role in clearing bacterial, viral, fungal, and parasitic infections [38]. When macrophages are stimulated, iNOS produces NO, which regulates blood pressure and has antibacterial activity. However, when the iNOS-derived NO is overproduced, it causes excessive expansion of blood vessels and tissue damage [39]. In this case, inducible haem oxygenase 1 (HO-1) plays a regulatory role as a rate-limiting enzyme; it catalyses the metabolism of haem into bilirubin, carbon monoxide (CO), and iron ions to regulate iNOS-mediated production of NO [40,41]. HO-1 and CO suppress the expression of iNOS and NO production in activated macrophages by deactivating nuclear factor-kappa B (NF-κB) [38,40,42]. Surprisingly, curcumin has a similar function; it indirectly inhibits NO production via inhibition of iNOS through suppression of NF-κB [36,43,44]. The NO-scavenging activity of curcumin and its derivatives, curcumin and THC, is potent, and the IC50 values are: curcumin ≈ THC > DMC > BDMC ≈ HHC > OHC [36]. Cyclooxygenase (COX) has two isozymes: COX-1 and COX-2. Its function is to catalyse the synthesis of prostaglandin from arachidonic acid. The role of COX-1 is to protect the gastric mucosa and maintain kidney function; the role of COX-2 is to promote prostacyclin (PGI2) production, which causes inflammation [45]. Curcumin, DMC, and BDMC are reported to inhibit COX-1 and COX-2 (32.0%, 38.5%, and 39.2% COX-1 inhibitory activity at a dose of 125 μg/mL; 89.7%, 82.0%, and 58.9% COX-2 inhibitory activity at a dose of 125 μg/mL, respectively). The COX-1 inhibitory effect of these compounds was slightly below that of the positive controls Aspirin (μg/mL), ibuprofen (2.06 μg/mL), and naproxen (2.52 μg/mL); however, they exhibited higher COX-II inhibitory activity than positive controls [45]. Furthermore, RAW264.7 macrophages treated with curcumin exhibited increased expression of HO-1 and decreased expression of iNOS protein and decreased iNOS promoter activity, which reduced NO production [38]. Moreover, curcumin can block lipopolysaccharide (LPS)-mediated expression of tumour necrosis factor α (TNF-α), interleukin (IL)-1β, and IL-6. Curcumin may alleviate inflammation through the nuclear factor-E2-related factor 2 (Nrf2)/HO-1 pathway [46]. Furthermore, the anti-inflammatory effect may be associated with activator protein-1 (AP-1) [47]. NF-κB and AP-1 are two transcription factor genes that are crucial to the LPS-induced inflammatory response and are commonly overexpressed in cancer cells [48]. In this situation, by inhibiting NF-κB, curcumin treatment results in the death of malignant cells and inhibits inflammation. Of note, one study suggested that the role of curcumin in the inflammatory system was like a pro-drug that requires oxidative activation to a reactive metabolite to exert anti-inflammatory effects [49]. Stably transfected RAW264.7 cells showed reduced luciferase activity expressed downstream of an NF-κB response element when treated with Curcuminoids and induced with LPS. Furthermore, NF-κB inhibition by curcumin and DMC involves the oxidation of these compounds into reactive electrophiles (Fig. 2) [50]. In contrast, BDMC is less likely to undergo spontaneous oxidative transformation due to the lack of ortho-methoxy groups that can accelerate the oxidation of the phenolic hydroxyl [51]. This also explains why curcumin and DMC have similar inhibitory pathways [36,45], while BDMC is different. In view of the lack of uncertainty of curcumin oxidative in vivo, the anti-inflammatory potential of oxidation-independent BDMC may be a new research direction.

Fig. 2.

Fig. 2

Mechanistic differences in the inhibition of NF-κB by curcumin, DMC, and BDMC.

3.3. Anti-diabetes

Diabetes is a chronic metabolic disease. According to the guidelines published by the American Diabetes Association in 2021, diabetes is diagnosed under one of the following conditions: glycosylated haemoglobin (HbA1c) −6.5%, fasting plasma glucose −126 mg/dL, or 2-h plasma glucose −200 mg/dL, or random blood glucose −200 mg/dL [52]. Diabetes can be categorised into four types: insulin-dependent type 1 diabetes, non-insulin-dependent type 2 diabetes, gestational diabetes, and diabetes caused by other factors. Among these types, type 2 diabetes accounts for most cases of diabetes. In addition, due to the functional loss of blood sugar utilisation, diabetes often causes dyslipidaemia, which promotes mitochondria to produce ROS and causes macrophages to produce pro-inflammatory factors, such as iNOS and ROS, eventually leading to complications, such as heart, kidney, and liver disease [53]. Incretin is a collective term for peptide hormones that act on pancreatic β cells to stimulate insulin secretion [54]. Research has discovered that curcumin can inhibit gluconeogenesis enzymes glucose-6-phosphatase (G6Pase), phosphoenolpyruvate carboxykinase (PEPUC), and adenosine 5′-monophosphate-activated protein (AMP) kinase activity to suppress liver gluconeogenesis and glycolysis. Moreover, curcumin can increase the effect of insulin in inhibiting glycolysis [55]. In an animal model, treatment with 15 mg/kg and 30 mg/kg curcumin significantly reduced blood sugar, vasoconstrictor pressure, proteinuria, polyuria, serum creatinine, and blood urea nitrogen (BUN) in male diabetic Sprague–Dawley rats; furthermore, it reduced the production of kidney lipid peroxidation products (malondialdehyde) and increased the activity of antioxidant enzymes glutathione, catalase, and superoxide dismutase [56]. Recent research highlighted that curcumin could significantly increase GLP-1 secretion in GLUTag cells [57]. The secretion of GLP-1 requires a β-diketone structure and an aromatic ring with at least one methoxy group; curcumin has a β-diketone structure with two methoxy groups, making it the most potent candidate. In contrast, BDMC, which lacks a methoxy group, and THC, which lacks the β-diketone structure, cannot stimulate GLP-1 secretion [57,58]. In a clinical trial, 240 subjects who met the pre-diabetes criteria were divided into control and placebo groups. After treatment with 1.5 g of curcuminoids daily for 12 months, there was a significant decrease in the subjects' diabetes indicators (glycated haemoglobin, fasting blood glucose, and glucose tolerance), improved pancreatic β-cell function, and increased performance of anti-inflammatory factor adiponectin [59]. Together, these data support the therapeutic potential of curcumin in controlling diabetes.

3.4. Anti-colorectal cancer

One of the basic concepts of cancer is the balance between cell proliferation and cell death [60]. When the apoptotic signals lose their function, cells proliferate out of control, leading to cancer [61]. Apoptotic signals are generated through two main pathways: in the intrinsic pathway, the mitochondrial membrane inhibits the expression of anti-apoptotic proteins Bcl-2 and Bcl-xL; in contrast, increasing the death receptors in the extrinsic apoptotic pathway triggers TNF-related apoptosis [62]. A study highlighted that curcumin disturbed the balance of mitochondrial membrane potential and increased the inhibition of Bcl-xL protein [63]. Furthermore, a study has revealed that curcumin significantly increases the expression of death receptor 5 (DR5) on both the mRNA and protein level [64]. Curcumin modulates tumour cell growth by regulating multiple cell signalling pathways. In addition to the cell survival pathway (Bcl-2, Bcl-xL) and death receptor pathway (DR4, DR5), there is the cell proliferation pathway (cyclin D1, c-myc), caspase activation pathway (caspase-8, -3, -9), tumour suppressor pathway (p53, p21), mitochondrial pathways, and protein kinase pathway (JNK, Akt, and AMPK) [65]. In the aforementioned pathways (Fig. 3), curcumin has shown significant anticancer effects in vitro and in vivo against several types of cancer, including prostate cancer, breast cancer, and colorectal cancer.

Fig. 3.

Fig. 3

Mechanism of induction of apoptosis by curcumin in colorectal cancer.

Colorectal (colon) cancer is one of the leading tumours globally. It is considered among the third most common cancers worldwide, with an array of high morbidity and mortality, being the fourth highest cause death [66,67]. Unfortunately, although treatment is mainly based on surgical resection, many patients continue to have a high risk of tumour recurrence [68]. COX-2 overexpression is observed in up to 90% of sporadic colon cancers and 40% of colon adenomas [69]. Therefore, specific COX-2 inhibitors have been clinically studied as agents for colon cancer chemoprevention; however, there are still doubts about their safety as they may increase cardiovascular risk [70,71]. Studies have shown that administration of curcumin can reduce the levels of oxidative DNA adduct 3-(2-deoxy-β-dierythro-pentafuranosyl)-pyr[1,2-α]-purin-10(3H) one (M1G) in malignant colorectal cells without changing the level of COX-2 protein (Table 1.) [72].

Table 1.

Effects of curcumin in colon cancer.

Model Curcumin dose/concentration Affected signalling pathways Mechanism Reference
Clinical trial 450, 1800, 3600 mg/day Curcumin reduces the adenoma burden in patients with colorectal cancer. M1G ↓
COX-2 -
[72]
In vivo
 Female F344 rats
2% mixed with daily diet Curcumin prevents colon cancer in rodent models. M1G ↓
COX-2 ↓
[73]
In vitro
 HCT116
10 μM Curcumin regulates miR-21, tumour growth, invasion, and metastasis of AP-1 binding ↓
miR-21 ↓
Pdcd4 ↑
[75]
In vitro
 HCT116
10 μM Curcumin induces cell cycle arrest and cellular senescence and down-regulates autophagosome ATG5 protein ↓
Cleavage of PARP ↑
Cell viability ↓
p53 ↑
[76]
In vivo
  Specific pathogen-free wild-type (WT) 129/SvEv mice Germ-freeIl10/mice
0.05% and 1% diets Curcumin reduced or eliminated colonic tumour burden in AOM-induced colon cancer. Tumour burden ↓
Colon weight/length ratio ↓
bacterial richness ↑
[78]
In vivo
 Swiss-Webster male mice
60 mg/kg Curcumin modulates polyamines synthesis, morphological changes, oxidative stress, and alters homeostasis and tumour development. Arginase Activity ↓
ACF formation ↓
Nitrotyrosine ↑
formation↓
Apoptosis ↑
HES-1 ↑
TGF-β ↑
Lipid peroxides ↓
[77]
In vivo
 HCT116 xenograft in nude mice
1 g/kg Curcumin targets NF-κB to improve the response of radiation therapy. Tumour regrowth ↓
Ki-67 proliferation index ↓
NF-κB activity ↓
[81]
In vitro
 HCT116
 HT29
 SW620
25 μM Curcumin inhibits the proliferation and post-irradiation clonogenic survival of multiple colo-rectal cancer cell lines. NF-kB activity ↓
BCL-xL ↓
BCL-1 ↓
Cyclin D1 ↓
COX-2 ↓
[82]
In vitro
 HCT116
20 μM Curcumin enhances anti-proliferation and induces apoptosis in 5-FU treatment. Caspase-8 ↑
Caspase-9 ↑
Caspase-3 ↑
Bax ↑
PARP ↑
BCL-xL ↓
Cyclin D1 ↓
[83]
In vitro
 HCT116
20 μM Curcumin enhances chemosensitivity in 5-FU treatment. Cell growth ↓
Colonspheres ↓
Apoptosis ↑
[84]

Unlike in clinical studies, both M1G and COX-2 protein are reduced in animal models following treatment with curcumin [73]. This outcome is hypothesised to be due to the ability of curcumin to inhibit COX-2, which is mainly caused by the reduction or conjugation of generated species [74]. In HCT 116 colorectal cancer cells treated with curcumin, the cell cycle was arrested in the G2/M phase via miR-21 gene regulation, which inhibited tumour tissue growth [75]. In another study, curcumin induced senescence and inhibited the growth of HCT116 colorectal cancer cells [76]. In an in vivo model, curcumin significantly reduced aberrant crypt foci (ACF) and iNOS expression and arginase activity in 1,2-dimethylhydrazine (DMH)-induced ACF mice [77]. Furthermore, a curcumin-supplemented diet increased survival, decreased colon weight/length ratio, and decreased tumour burden in rats with AOM-induced colon cancer [78]. Although curcumin seems effective in preventing or treating colon cancer in vivo and in vitro, oral administration of curcumin results in its rapid metabolism, and approximately 60–70% of the compound is excreted in the faeces [67]. In clinical trials, quantifiable serum levels are not achieved until high doses are administered [69]. This observation can be attributed to the low bioavailability of curcumin. Because of the limited clinical effects of curcumin alone [79], current studies tends to increase the bioavailability of curcumin or administer it as an adjuvant treatment [80]. In an in vivo study in HCT116 xenograft nude mice, curcumin targeted NF-κB and improved the response of radiation therapy to colorectal cancer [81]. Another in vitro model study confirmed that curcumin prevented the proliferation and post-irradiation clonogenic survival of multiple colorectal cancer cell lines by suppressing radiation-induced NF-κB activation [82]. In addition to radiation therapy, curcumin combined with chemotherapy has been extensively studied. In a study evaluating a combination treatment regimen of 5-fluorouracil (5-FU) and curcumin in colorectal cancer cells, cell cycle analysis revealed that treatment with curcumin and 5-FU led to accumulation of colorectal cancer cells in the S cell cycle phase and induction of apoptosis [83]. In another study, when compared with treatment with 5-FU alone, pre-treatment with curcumin significantly enhanced the effect of 5-FU on colorectal cancer cells [84].

3.5. Other physiological activities

Curcumin has potential in the treatment of a variety of cancers in addition to colorectal cancer. Prostate cancer is the second most commonly diagnosed cancer in men, and ranks fifth as the leading cause of death globally [85]. The current mainstream treatment methods of localised and androgen-dependent prostate cancer (ADPC) include hormonal treatment, surgery, and radiotherapy. However, these cancerous cells progress to androgen-independent prostate cancer (AIPC) over time and are no longer dependent on hormones [86]. Research has shown that curcumin inhibits the proliferation of prostate cancer cells and induces apoptosis [87] by interfering with several cellular pathways, including mitogen-activated protein kinase (MAPK), epidermal growth factor receptor (EGFR), and NF-κB [86,88]. The dilemmas faced in the treatment of breast cancer and prostate cancer are similar. After lumpectomy, radiotherapy, chemotherapy, and endocrine therapy, the recurrence rate of breast cancer remains high [89]. A study evaluated the effects of curcumin on cell cycle regulatory proteins, matrix metalloproteinases (MMPs), and NF-κB in MDA-MB-231 and BT-483 breast cancer cells. The results indicated that curcumin exhibited antiproliferative activity by downregulating NF-κB [90]. Furthermore, curcumin has in vivo and in vitro effects in head and neck squamous cell carcinoma [91,92] and ovarian cancer through similar pathways [93,94].

In addition to the aforementioned functionalities, curcumin can reduce mouse oxidized protein, cytokine IL-1β, astrocyte skeleton protein glial fibrillary acidic protein (GFAP), insoluble and soluble amyloid β-amyloid expression, and inhibit microgliosis to prevent Alzheimer's disease [95]. Furthermore, curcumin has in vivo antibacterial activity that can prevent and restore damage to the stomach caused by Helicobacter pylori [96]. Recent studies have reported the antiparasitic properties of curcuminoids (DMC and BDMC), which inhibited TGR activity, giving them the ability to fight Taenia crassiceps cysticercosis. Moreover, curcuminoids (including curcumin, DMC, and BDMC) can effectively inhibit neuraminidase activity to inhibit influenza viruses H1N1 and H9N2. Of note, several studies have stated that curcumin has the potential to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [97,98]. In addition, studies have mentioned that curcumin can inhibit autoimmune deficiency syndrome (AIDS), Parkinson's disease, allergies, cardiovascular diseases, and other physiological activities [11,99102].

4. Application dilemma

Although curcumin has been shown to exhibit therapeutic and protective effects in various diseases, studies have shown that oral administration of 8 g/day of curcumin will only results in a plasma concentration of about 2.5 ng/mL [103]. This is due to the low water solubility and bioavailability of curcumin, and it is also degraded by the digestive system or converted into metabolites [104]. In general, higher doses may be more effective, but further consideration should be given to potential side effects.

Turmeric and curcumin have been used for decades as food additives, supplements, and medicines. According to reports by the Joint United Nations and World Health Organization Expert Committee on Food Additives (JECFA) and European Food Safety Authority (EFSA), the ADI of curcumin is 0–3 mg/kg body weight [10,105,106]. A long-term curcumin carcinogenicity study by the National Toxicology Program (NTP) revealed that the incidence of malignant neoplastic lesions (carcinomas) did not reach statistical significance and the observed effects were not dose-dependent, were in agreement with historical control values, and were not consistent across sexes and/or species; these observations eliminated concerns of genotoxicity [107]. Nevertheless, there are still some records of side effects in previous research. For example, diarrhoea, headache, rash, and yellow stool have been reported after oral administration of 500–12000 mg of curcumin; however, the same study stated that a daily intake of up to 12000 mg of curcumin has no harmful effects on individuals [108]. In another study, dose-limiting toxicity was not observed up to 3600 mg of curcumin. Still, patients reported two types of gastrointestinal adverse events, and a rise in serum alkaline phosphatase levels was also observed [109]. These concerns make administering high doses of curcumin less optimal. At present, the most studied strategy is the modification of curcumin and its delivery systems, using nanoparticles [110112], micellization [113115], and chemical conjugation [104,116118]. The application of these technologies can improve the stability, solubility, in vivo absorption, biological activity, and safety of curcumin without increasing the dosage. Recent studies have begun to explore more strategies that can help improve the application of curcumin.

5. Novel application strategies

5.1. Curcumin analogues and related substances

The main functionalities of curcumin are derived from three reactive functional groups on the chemical structure, including one diketone moiety and two phenolic groups. These groups determine the biological activity of curcumin, including hydrogen donation reactions, reversible and irreversible nucleophilic addition reactions, hydrolysis, degradation, and enzymatic reactions [27]. Analogues or degradation products that are different from curcumin can also promote or reduce its functionality. Therefore, direct supplementation of curcumin analogues or degradation products may be a potential strategy to improve its effectiveness (Table 2).

Table 2.

Activities of curcumin analogues, metabolites, and degradation products.

Curcumin analogues/related substances Bioactivity Reference
Demethoxycurcumin (DMC) • Suppressed migration and invasion in MDA-MB-231 human breast cancer cell line. [120]
• Induced Bcl-2-mediated G2/M arrest and apoptosis in human glioma U87 cells. [121]
• Exerted a cytostatic effect at G2/M in MCF-7 human breast tumour cells. [122]
• Inhibited cell proliferation and induced apoptosis in HER2-overexpressing bladder cancer cells. [125]
• Increased the sensitivity of cisplatin-resistant cancer cells. [124]
• The inhibition and degradation activity of bacteria. [126,127]
Bisdemethoxycurcumin (BDMC) • Cytotoxic against human ovarian cancer OVCAR-3 cells. [128]
• Prevented DMH-induced colon carcinogenesis. [129]
• Accelerated gastric ulcer healing. [130]
• Induced apoptotic cell death in Hep 3B cells. [131]
• Inhibited MCF-7 breast cancer cell proliferation. [132]
• Inhibited adipogenesis in 3T3-L1 preadipocytes and suppressed obesity. [133]
• Used as an antibacterial agent to relieve antibiotic resistance. [134]
Tetrahydrocurcumin (THC) • Ameliorated oxidative stress-induced renal injury. [139]
• Alleviated the oxidative stress caused by cholesterol intake. [140]
• Antioxidant and vascular protective effects in L-NAME-induced hypertension. [141]
• Ameliorated insulin resistance in fatty acid-induced hepatic steatosis. [142]
• Preventative effects on azoxymethane-induced colon carcinogenesis. [143]
• Anti-angiogenic effects on implanted hepatocellular carcinoma. [144]
Ferulic acid • Improved cardiovascular and kidney structure and function in hypertensive rats. [148]
• Attenuated acute renal injury induced by cisplatin. [149]
• Improved cognitive function. [150152]
• Inhibited cell proliferation and invasion in HeLa and CaSki cervical cancer cells. [153]
Vanillin • Induced G0/G1 arrest and apoptosis in human HT-29 colon cancer cells. [156]
• Induced apoptosis in human hepatic carcinoma HepG2 and neuroblastoma SH-SY5Y cells. [157]
• Reduced apoptosis and exerted neuroprotective effects in rats with spinal cord injury. [158]
• Promoted early neurofunctional development in neonatal rats. [159]
Calebin A • Protected cells from beta-amyloid insult. [161]
• Inhibited cell growth and induced apoptosis in drug-resistant human gastric cells. [162]
• Regulated survival and inflammatory gene products, leading to inhibition of cell growth and chemosensitisation. [160]
• Suppressed NF-κB-mediated proliferation, invasion, and metastasis of human colorectal cancer cells. [163]

5.1.1. Curcumin analogues

DMC and BDMC are the most abundant curcuminoids after curcumin, and research has shown that their functions may be better than curcumin [119]. For example, in the MDA-MB-231 human breast cancer cell line, DMC exhibited anti-invasive activity by modulating the expression of invasion-associated proteins [120]. In human glioma U87 cells, DMC bound more efficiently to the Bcl-2 putative active site and induced Bcl-2-mediated G2/M arrest and apoptosis [121]; similar results were observed in MCF-7 cells [122]. In HER2-overexpressing bladder cancer cells, it significantly suppressed the expression of HER2, preferentially inhibited cell proliferation, and induced apoptosis. In human cervical cancer HeLa cells, it suppressed migration and invasion via inhibition of NF-κB pathways [123]. Furthermore, some studies reported that DMC increased the sensitivity of cisplatin-resistant cancer cells, such as A549 human alveolar basal epithelial adenocarcinoma cells [124] and HER2-overexpressing bladder cancer cells [125]. In addition to its antagonistic effects on specific cancer cells, DMC has potential as an antimicrobial against pathogens such as Candida albicans ATCC 10231 [126] and methicillin-resist Staphylococcus aureus [127].

Compared with DMC, BDMC has two fewer methyl groups, which means it has better water solubility and bioavailability. The structure of BDMC is responsible for its loss of antioxidant activity [36]; however, it has an increased anti-inflammatory effect [50]. However, there appears to be a trend in other specific conditions. In human ovarian cancer OVCAR-3 cells, BDMC exhibited higher cytotoxic activity than curcumin and DMC against ovarian cancer cells [128]. In a DMH-induced carcinogenesis in vivo model, intragastric BDMC significantly reduced the number and size of tumours in the colon in addition to hepatic oxidative stress [129]. In a gastric ulcer model, BDMC suppressed iNOS-mediated inflammation and directly accelerated gastric ulcer healing [130]. In human liver cancer Hep3B cells, BDMC decreased cell viability and induced S phase arrest, DNA damage, and cell apoptosis [131]. In human breast cancer MCF-7 cells, BDMC increased the concentration of ROS in the cells, inhibiting cancer cell proliferation [132]. In addition to its anti-cancer effects, BDMC has anti-obesity effects; it inhibited adipogenesis in 3T3-L1 preadipocytes and suppressed obesity in a high-fat-diet-induced in vivo model [133]. Finally, BDMC has antimicrobial effects and may be a potential natural antibacterial agent to ameliorate antibiotic resistance [134]. DMC and BDMC are potent and are comparable to curcumin in many studies; however, their properties are more anti-inflammatory than purely anti-oxidative due to chemical structural factors, and both have better water solubility and bioavailability than curcumin [135]. These two compounds can be used as alternative strategies where curcumin fails.

THC is one of the primary metabolites of curcumin; it lacks an α,β-unsaturated carbonyl moiety [136] and has different spectral properties from curcumin, usually producing white crystals at room temperature. Due to its higher bioavailability and different biological activity and molecular mechanisms than curcumin, some studies suggest that it has more potential for development than curcumin [137]. THC is well known for its anti-oxidative activity. During the anti-oxidative process, the β-iketone moiety C–C bond will be cleaved and exhibit anti-oxidative activity [138]. THC can be used against ferric nitrilotriacetate (Fe-NTA)-induced oxidative renal damage in male ddY mice [139], and it can effectively alleviate oxidative stress in cholesterol-fed rabbits [140]. Furthermore, THC has shown antioxidative and vascular protective effects in L-NAME-induced hypertension in rats [141]. In fatty acid-induced hepatic steatosis, THC ameliorated insulin resistance in HepG2 cells [142]. Some studies have shown that THC has partial anticancer effects, such as preventing azoxymethane-induced colon carcinogenesis [143], and anti-angiogenic effects on implanted hepatocellular carcinoma in nude mice [144]. Overall, THC is more effective than curcumin in oxidative stress-related diseases due to its excellent antioxidant effect. Overall, the chemical structure of THC enhances its antioxidant effects; therefore, it can be more effective in oxidative stress-related diseases.

5.1.2. Curcumin related substances

Ferulic acid and vanillin were structurally identified as curcumin-derived radical reaction products [145], and both have higher water solubility as curcumin degradation products [146]. Although studies have highlighted that the two compounds may not be major curcumin degradation products [147], as long-studied and biologically active phenolic compounds, they may still retain some functional properties of turmeric after degradation. Research has confirmed that ferulic acid improves cardiovascular and kidney structure and function in hypertensive rats [148] and has the ability to attenuate acute renal injury induced by cisplatin [149]. Furthermore, studies have shown that ferulic acid improves cognitive function [150152]. Regarding anti-cancer activity, ferulic acid can significantly inhibit cell proliferation and invasion in HeLa and CaSki, two cervical cancer cells [153]. Ferulic acid may not possess as effective biological activity as curcumin, but its use as an adjunct to curcumin can compensate for its mechanical deficiencies [150].

Although vanillin and ferulic acid have similar structures, vanillin is more remarkable in food applications. Like curcumin, it is a common flavour additive and is probably the most widely used flavouring agent for sweet foods. Furthermore, its antioxidant activity helps stabilise the oxidation and degradation of food components [154]. Vanillin and ferulic acid have similar biological activity, including antioxidant, anti-inflammatory, neuroprotective, and anticancer properties [155]. In human colon cancer HT-29 cells, vanillin arrests the cell cycle in G0/G1 phase and significantly increases apoptosis in the sub-G0 phase [156]; this effect has also been reported in human hepatic carcinoma and neuroblastoma cells [157]. Recent studies have found that, in addition to its neuroprotective effects [158], vanillin promotes early neurological development and improves hypoxic-ischaemic brain damage in neonatal rats [159]. More notably, due to its lack of toxicity in rats [162], vanillin is well-suited for curcumin application strategies.

Calebin A (4-[3-methoxy-4-hydroxyphenyl]-2-oxo-3-enebutanyl-3-[3-methoxy-4-hydroxyphenyl] propenoate) is a compound that had not been isolated or identified in turmeric until recently [160]. It was originally identified to protect cells from beta-amyloid insult [161]. Follow-up studies found that it can inhibit cell growth and induce apoptosis in drug-resistant human gastric carcinoma MDR cell line SGC7901/VINCRISTINE. It resulted in a reduction in S phase and G2/M phase arrest and modulated the activity of MAPK family members [162]. Studies have reported that calebin A inhibits the NF-κB activation pathway via interaction with p65 and enhances multiple cancer cell apoptosis [160]. In addition, one study reported that calebin A had anti-proinflammatory and anti-tumour activity in TNF-β-stimulated colorectal cancer cells [163] and enhanced the effect of the anticancer drug 5-FU [164]. Despite the sparsity of research on calebin A, according to the current literature, it has potential in cancer prevention and treatment.

In conclusion, the strategy of applying curcumin analogues and related substances to replace curcumin is based on its structural activity. Ideally, it increases bioavailability, leaving the active structure unchanged. According to the reported results, curcumin analogues and related substances have many different efficacy aspects and application potential.

5.2. Gut microbiota regulation

The gut microbiota is an entire population of microorganisms located in the gut [165]. It is associated with a variety of human diseases, including intestinal disorders, such as inflammatory bowel disease (IBD) [166] and irritable bowel syndrome (IBS) [167], and metabolic diseases, such as obesity and diabetes [168170]. The bioavailability of curcumin is extremely poor; however, studies have pointed out that with administered orally, curcumin has preferential distribution and accumulation in the intestinal tract and can reach the level of biological activity [171]. In the intestinal tract, curcumin has a regulatory impact on the gut microbiota, influencing microbial abundance, variety, and composition [172]. Curcumin significantly increases the proportion of beneficial microbiota relative to pathogenic microbiota by increasing the abundance of Bifidobacterium, Lactobacillus, and butyrate-producing bacteria and reducing the abundance of Prevotellaceae, Coriobacteriaceae, enterobacteria, and Enterococcus. In addition to its anti-inflammatory and anti-colorectal cancer activity, these changes in the intestinal microbiota can explain the immunomodulatory and anti-hyperlipidaemic effects of curcumin [173]. Furthermore, the reduction of Prevotellaceae, Bacteroidaceae, and Rikenellaceae, which are often linked to the onset of systemic diseases [172], demonstrates the potential of curcumin as a dietary supplement. One study reported that curcumin dramatically shifted the overall structure of a high-fat diet, disrupted gut microbiota towards that of lean rats fed a normal diet, and altered the gut microbial composition [174]. Some studies suggest that this can be attributed to the curcumin promotes a shift from pathogenic to beneficial bacterial strains and further affect intestinal metabolisms, such as fatty acids [175,176] and bile acid [176,177].

In addition to directly regulating the intestinal microbiota, leading to changes in microbial richness, diversity, and composition, curcumin is bio-transformed into various metabolites, such as dihydrocurcumin (DHC), THC, and ferulic acid by intestinal microbiota via demethylation, hydroxylation, demethoxylation, and decomposition [175,178]. For example, studies have shown that Escherichia coli from the intestinal tract will sequentially convert curcumin into DHC and THC [179]. Take the THC mentioned in Section 5.1.1 as an example, the amount of THC and its conjugates (as sulfates and glucuronides) were higher in the liver and serum after dietary administration of either curcumin or THC compared to the amount of curcumin and its conjugates. Therefore, THC is more readily absorbed from the gastrointestinal tract than curcumin [139]. In addition to this, some studies have reported that many of these metabolites are more biologically active and bioavailable than curcumin [175,180,181]. The above research note that curcumin not only achieves health benefits by regulating gut microbiota, but also by being metabolized by these microorganisms to produce bioavailable metabolites with similar effects to curcumin, thus increasing health benefits [173].

5.3. Exosome vesicles

Previous studies on improving curcumin bioavailability have focused on chemical or microbial modification, binding phospholipids to form complexes, entrapment using liposomes, and nanoparticles as carriers [80,104,119,182184]. Exosomes are extracellular microvesicles with a particle size between 30 nm and 150 nm that carry a large number of proteins, lipids, RNA, and DNA and can be used as intercellular messaging tools [182]. Because exosomes have the ability to shuttle in and out of cells, the use of exosomes as nano-drug carriers has potential for new therapeutic applications [185]. In addition, many studies have suggested that exosomes as drug carriers have the potential to overcome the technology-related limitations of traditional nanoparticles [186]. For example, exosomes have a longer circulating half-life, are more easily internalised by cells, and can be linked to one or more tumour-recognition ligands to enhance their targeting capabilities, which make exosomes ideal nanoparticle drug delivery vesicles [187]. Many studies have used exosomes to coat curcumin and deliver it to targeted cells via membrane fusion [182,188].

The coating of functional components with exosomes can be divided into two methods: active and passive. The passive method only requires co-culture with functional components and cells or purification of exosomes from the culture medium of the cell culture and mixing them so that functional components can diffuse into exosomes via concentration difference (Fig. 4) [189]. In 2010, a study combined exosomes from mouse lymphoma with curcumin to produce curcumin-coated exosomes [182]. Subsequent studies have confirmed that curcumin treatment of exosomes produced by chronic myelogenous leukaemia (CML) can attenuate their ability to promote angiogenesis and regulate endothelial barrier tissue, thereby affecting tumour progression [190]. Another study used curcumin to intervene in mouse brain endothelial cells (MBECs) to produce curcumin-containing exosomes. It was confirmed that curcumin could ameliorate oxidative stress during endothelial cell damage and regulate tightness. Expression of connexins (ZO-1, claudin-5, and occludin) and adhesion junction protein (VE-cadherin) and increase endothelial cell permeability [191]. In contrast, the active method is more complicated and involves techniques such as ultrasound, extrusion, membrane perforation, or repeated freezing and thawing [192].

Fig. 4.

Fig. 4

Curcumin-loaded exosome vesicle preparation and its advantages.

Studies have shown that curcumin coating of exosomes effectively increases the concentration of curcumin in the plasma and improve bioavailability. Injection of 100 mg/kg body weight of curcumin in exosomes can reach the highest plasma concentration in 30 min (1250 ng/mL), which is 5–10-fold that of curcumin alone [182]. Other studies have confirmed that exosome coating effectively improves the solubility, stability, and bioavailability of curcumin [183,184]. In recent years, the exosome drug delivery system has been an emerging research field that is a promising and novel concept. Existing literature clarifies that exosome-coated curcumin has anti-inflammatory [182,184], endothelial protective [183,191], neuroprotective [193], and anti-cancer properties [194]. Turmeric-derived exosome-like vesicle-related research has begun to attract attention. Turmeric-derived nanoparticles (TDNPs) or turmeric-derived nanovesicles (TNVs) isolated from turmeric can effectively alleviate colitis. Research indicates that oral administration of TDNPs prevents colitis and promotes wound repair in colitis [195]. Meanwhile, another study reported that oral administration of TNVs restored the damaged gut barrier, modulated gut microbiota, reshaped the macrophage phenotype, then increased its anti-inflammatory effect [196]. Compared with nanoparticle delivery systems, TDNPs and TNVs are natural colon-targeting therapeutics that have the advantages of low toxicity and ease of large-scale production. Although there are very few related studies, this is a topic with great potential for future research based on the current results.

6. Conclusion

Curcumin, the active component of C. longa extract, has been extensively studied in recent decades. These studies have confirmed its antioxidant, anti-inflammatory, anti-diabetes, and anticancer effects. However, the application of curcumin has been restricted by its low water solubility, which results in low cellular uptake, poor oral bioavailability, and low chemical stability. These factors make its clinical effectiveness and in vivo efficacy lower than its in vitro activity. Structural modification, synergistic combination therapy, and drug delivery systems are currently the most common solutions that are proven to increase the bioavailability of curcumin and improve its effectiveness. With the development of innovative technologies, there are an increasing number of strategies to solve this problem. For example, after passing through the digestive system and undergoing microbial metabolism, curcumin may be metabolised into a more biologically active form, thereby affecting overall body functions and organs. Therefore, directly involving these metabolites or degradants may achieve more direct effects. In addition, curcuminoids and curcumin analogues provide similar biological activity but higher bioavailability because of their structural similarity to curcumin. We can also take advantage of the low bioavailability of curcumin and allow it to directly regulate gut microbiota, leading to changes in microbial richness, diversity, and composition. Finally, the application of an exosome vesicle delivery system, greatly improves the bioavailability of curcumin and provides the possibility of targeted therapy. Overall, these successful findings provide valuable information for the future study of curcumin. Furthermore, the potential function and development of curcumin should be established.

Acknowledgements

This study was supported by the Ministry of Science and Technology [109-2320-B-002 -012 -MY3 and 110-2320-B-002 -019 -MY3].

Funding Statement

This study was supported by the Ministry of Science and Technology [109-2320-B-002 -012 -MY3 and 110-2320-B-002 -019 -MY3].

References

  • 1. Chattopadhyay I, Biswas K, Bandyopadhyay U, Banerjee RK. Turmeric and curcumin: biological actions and medicinal applications. Curr Sci. 2004:44–53. [Google Scholar]
  • 2. Amalraj A, Pius A, Gopi S, Gopi S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives–A review. J Tradit Complement Med. 2017;7:205–33. doi: 10.1016/j.jtcme.2016.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Jayaprakasha GK, Jena BS, Negi PS, Sakariah KK. Evaluation of antioxidant activities and antimutagenicity of turmeric oil: a byproduct from curcumin production. Z Naturforsch C Biosci. 2002;57:828–35. doi: 10.1515/znc-2002-9-1013. [DOI] [PubMed] [Google Scholar]
  • 4. Lantz R, Chen G, Solyom A, Jolad S, Timmermann B. The effect of turmeric extracts on inflammatory mediator production. Phytomedicine. 2005;12:445–52. doi: 10.1016/j.phymed.2003.12.011. [DOI] [PubMed] [Google Scholar]
  • 5. Tilak JC, Banerjee M, Mohan H, Devasagayam T. Antioxidant availability of turmeric in relation to its medicinal and culinary uses. Phytother Res. 2004;18:798–804. doi: 10.1002/ptr.1553. [DOI] [PubMed] [Google Scholar]
  • 6. Naz S, Jabeen S, Ilyas S, Manzoor F, Aslam F, Ali A. Antibacterial activity of Curcuma longa varieties against different strains of bacteria. Pakistan J Bot. 2010;42:455–62. [Google Scholar]
  • 7. Aggarwal BB, Yuan W, Li S, Gupta SC. Curcumin-free turmeric exhibits anti-inflammatory and anticancer activities: identification of novel components of turmeric. Mol Nutr Food Res. 2013;57:1529–42. doi: 10.1002/mnfr.201200838. [DOI] [PubMed] [Google Scholar]
  • 8. Li M, Yue GG-L, Tsui SK-W, Fung K-P, Bik-San Lau C. Turmeric extract, with absorbable curcumin, has potent anti-metastatic effect in vitro and in vivo. Phytomedicine. 2018;46:131–41. doi: 10.1016/j.phymed.2018.03.065. [DOI] [PubMed] [Google Scholar]
  • 9. Lima CF, Pereira-Wilson C, Rattan SI. Curcumin induces heme oxygenase-1 in normal human skin fibroblasts through redox signaling: relevance for anti-aging intervention. Mol Nutr Food Res. 2011;55:430–42. doi: 10.1002/mnfr.201000221. [DOI] [PubMed] [Google Scholar]
  • 10.JECFA. Sixty-first report of the Joint FAO/WHO Expert committee on food additives. 2004. [Google Scholar]
  • 11. Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol. 2009;41:40–59. doi: 10.1016/j.biocel.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. López-Lázaro M. Anticancer and carcinogenic properties of curcumin: considerations for its clinical development as a cancer chemopreventive and chemotherapeutic agent. Mol Nutr Food Res. 2008;52:S103–27. doi: 10.1002/mnfr.200700238. [DOI] [PubMed] [Google Scholar]
  • 13.Latruffe N. Natural products and inflammation. Multidisciplinary Digital Publishing Institute; 2017. [Google Scholar]
  • 14. Li R, Xiang C, Ye M, Li H-F, Zhang X, Guo D-A. Qualitative and quantitative analysis of curcuminoids in herbal medicines derived from Curcuma species. Food Chem. 2011;126:1890–5. doi: 10.1016/j.foodchem.2010.12.014. [DOI] [PubMed] [Google Scholar]
  • 15. Li H-X, Zhang H-L, Zhang N, Wang N, Yang Y, Zhang Z-Z. Isolation of three curcuminoids for stability and simultaneous determination of only using one single standard substance in turmeric colour principles by HPLC with ternary gradient system. LWT–Food Sci Technol. 2014;57:446–51. [Google Scholar]
  • 16. Poudel A, Pandey J, Lee H-K. Geographical discrimination in curcuminoids content of turmeric assessed by rapid UPLC-DAD validated analytical method. Molecules. 2019;24:1805. doi: 10.3390/molecules24091805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Ashraf K, Mujeeb M, Ahmad A, Ahmad N, Amir M. Determination of curcuminoids in curcuma longa linn. By UPLC/Q-TOF–MS: an application in turmeric cultivation. J Chromatogr Sci. 2015;53:1346–52. doi: 10.1093/chromsci/bmv023. [DOI] [PubMed] [Google Scholar]
  • 18. Osorio-Tobón JF, Carvalho PI, Barbero GF, Nogueira GC, Rostagno MA, de Almeida Meireles MA. Fast analysis of curcuminoids from turmeric (Curcuma longa L.) by high-performance liquid chromatography using a fused-core column. Food Chem. 2016;200:167–74. doi: 10.1016/j.foodchem.2016.01.021. [DOI] [PubMed] [Google Scholar]
  • 19. Erpina E, Rafi M, Darusman LK, Vitasari A, Putra BR, Rohaeti E. Simultaneous quantification of curcuminoids and xanthorrhizol in Curcuma xanthorrhiza by high-performance liquid chromatography. J Liq Chromatogr Relat Technol. 2017;40:635–9. [Google Scholar]
  • 20.(Taiwan) MoHaW. Taiwan Herbal Pharmacopeia (IV); 2021. [Google Scholar]
  • 21. Commission CP. The Pharmacopoeia of the People's Republic of China 2020 edition. 2020 [Google Scholar]
  • 22. Jiang T, Ghosh R, Charcosset C. Extraction, purification and applications of curcumin from plant materials-A comprehensive review. Trends Food Sci Technol. 2021;112:419–30. [Google Scholar]
  • 23. Zhu J, Sanidad KZ, Sukamtoh E, Zhang G. Potential roles of chemical degradation in the biological activities of curcumin. Food Funct. 2017;8:907–14. doi: 10.1039/c6fo01770c. [DOI] [PubMed] [Google Scholar]
  • 24. Somparn P, Phisalaphong C, Nakornchai S, Unchern S, Morales NP. Comparative antioxidant activities of curcumin and its demethoxy and hydrogenated derivatives. Biol Pharm Bull. 2007;30:74–8. doi: 10.1248/bpb.30.74. [DOI] [PubMed] [Google Scholar]
  • 25. Jankun J, Wyganowska-Świątkowska M, Dettlaff K, Jelińska A, Surdacka A, Wątróbska-Świetlikowska D, et al. Determining whether curcumin degradation/condensation is actually bioactivation. Int J Mol Med. 2016;37:1151–8. doi: 10.3892/ijmm.2016.2524. [DOI] [PubMed] [Google Scholar]
  • 26. Pabon H. A synthesis of curcumin and related compounds. Recl Trav Chim Pays-Bas. 1964;83:379–86. [Google Scholar]
  • 27. Priyadarsini KI. The chemistry of curcumin: from extraction to therapeutic agent. Molecules. 2014;19:20091–112. doi: 10.3390/molecules191220091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Lampe V, Milobedzka J. Studien über curcumin. Ber Dtsch Chem Ges. 1913;46:2235–40. [Google Scholar]
  • 29. Rao EV, Sudheer P. Revisiting curcumin chemistry part I: a new strategy for the synthesis of curcuminoids. Indian J Pharm Sci. 2011;73:262–70. doi: 10.4103/0250-474X.93508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Venkateswarlu S, Ramachandra MS, Subbaraju GV. Synthesis and biological evaluation of poly-hydroxycurcuminoids. Bioorg Med Chem. 2005;13:6374–80. doi: 10.1016/j.bmc.2005.06.050. [DOI] [PubMed] [Google Scholar]
  • 31. Barros L, Ferreira M-J, Queiros B, Ferreira IC, Baptista P. Total phenols, ascorbic acid, β-carotene and lycopene in Portuguese wild edible mushrooms and their antioxidant activities. Food Chem. 2007;103:413–9. [Google Scholar]
  • 32. Gülçin İ. Antioxidant and antiradical activities of L-carnitine. Life Sci. 2006;78:803–11. doi: 10.1016/j.lfs.2005.05.103. [DOI] [PubMed] [Google Scholar]
  • 33. Ak T, Gülçin İ. Antioxidant and radical scavenging properties of curcumin. Chem Biol Interact. 2008;174:27–37. doi: 10.1016/j.cbi.2008.05.003. [DOI] [PubMed] [Google Scholar]
  • 34. Halliwell B, Gutteridge JM. [1] Role of free radicals and catalytic metal ions in human disease: an overview. Methods Enzymol. 1990;186:1–85. doi: 10.1016/0076-6879(90)86093-b. [DOI] [PubMed] [Google Scholar]
  • 35. Wright JS. Predicting the antioxidant activity of curcumin and curcuminoids. J Mol Struct THEOCHEM. 2002;591:207–17. [Google Scholar]
  • 36. Morales NP, Sirijaroonwong S, Yamanont P, Phisalaphong C. Electron paramagnetic resonance study of the free radical scavenging capacity of curcumin and its demethoxy and hydrogenated derivatives. Biol Pharm Bull. 2015;38:1478–83. doi: 10.1248/bpb.b15-00209. [DOI] [PubMed] [Google Scholar]
  • 37. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. 2001;357:593–615. doi: 10.1042/0264-6021:3570593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Kim KM, Pae H-O, Zhung M, Ha H-Y, Ha YA, Chai K-Y, et al. Involvement of anti-inflammatory heme oxygenase-1 in the inhibitory effect of curcumin on the expression of pro-inflammatory inducible nitric oxide synthase in RAW264. 7 macrophages. Biomed Pharmacother. 2008;62:630–6. doi: 10.1016/j.biopha.2008.01.008. [DOI] [PubMed] [Google Scholar]
  • 39. Chung H-T, Pae H-O, Choi B-M, Billiar TR, Kim Y-M. Nitric oxide as a bioregulator of apoptosis. Biochem Biophys Res Commun. 2001;282:1075–9. doi: 10.1006/bbrc.2001.4670. [DOI] [PubMed] [Google Scholar]
  • 40. Oh G-S, Pae H-O, Lee B-S, Kim B-N, Kim J-M, Kim H-R, et al. Hydrogen sulfide inhibits nitric oxide production and nuclear factor-κB via heme oxygenase-1 expression in RAW264. 7 macrophages stimulated with lipopolysaccharide. Free Radic Biol Med. 2006;41:106–19. doi: 10.1016/j.freeradbiomed.2006.03.021. [DOI] [PubMed] [Google Scholar]
  • 41. Willoughby D, Moore A, Colville-Nash P, Gilroy D. Resolution of inflammation. Int J Immunopharmacol. 2000;22:1131–5. doi: 10.1016/s0192-0561(00)00064-3. [DOI] [PubMed] [Google Scholar]
  • 42. Oh G-S, Pae H-O, Choi B-M, Chae S-C, Lee H-S, Ryu D-G, et al. 3-Hydroxyanthranilic acid, one of metabolites of tryptophan via indoleamine 2, 3-dioxygenase pathway, suppresses inducible nitric oxide synthase expression by enhancing heme oxygenase-1 expression. Biochem Biophys Res Commun. 2004;320:1156–62. doi: 10.1016/j.bbrc.2004.06.061. [DOI] [PubMed] [Google Scholar]
  • 43. Manikandan R, Beulaja M, Thiagarajan R, Priyadarsini A, Saravanan R, Arumugam M. Ameliorative effects of curcumin against renal injuries mediated by inducible nitric oxide synthase and nuclear factor kappa B during gentamicin-induced toxicity in Wistar rats. Eur J Pharmacol. 2011;670:578–85. doi: 10.1016/j.ejphar.2011.08.037. [DOI] [PubMed] [Google Scholar]
  • 44. Jung KK, Lee HS, Cho JY, Shin WC, Rhee MH, Kim TG, et al. Inhibitory effect of curcumin on nitric oxide production from lipopolysaccharide-activated primary microglia. Life Sci. 2006;79:2022–31. doi: 10.1016/j.lfs.2006.06.048. [DOI] [PubMed] [Google Scholar]
  • 45. Ramsewak R, DeWitt D, Nair M. Cytotoxicity, antioxidant and anti-inflammatory activities of curcumins I–III from Curcuma longa. Phytomedicine. 2000;7:303–8. doi: 10.1016/S0944-7113(00)80048-3. [DOI] [PubMed] [Google Scholar]
  • 46. Liu L, Shang Y, Li M, Han X, Wang J, Wang J. Curcumin ameliorates asthmatic airway inflammation by activating nuclear factor-E2-related factor 2/haem oxygenase (HO)-1 signalling pathway. Clin Exp Pharmacol Physiol. 2015;42:520–9. doi: 10.1111/1440-1681.12384. [DOI] [PubMed] [Google Scholar]
  • 47. Divya CS, Pillai MR. Antitumor action of curcumin in human papillomavirus associated cells involves downregulation of viral oncogenes, prevention of NFkB and AP-1 translocation, and modulation of apoptosis. Mol Carcinog. 2006;45:320–32. doi: 10.1002/mc.20170. [DOI] [PubMed] [Google Scholar]
  • 48. Perrone D, Ardito F, Giannatempo G, Dioguardi M, Troiano G, Lo Russo L, et al. Biological and therapeutic activities, and anticancer properties of curcumin. Exp Ther Med. 2015;10:1615–23. doi: 10.3892/etm.2015.2749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Edwards RL, Luis PB, Varuzza PV, Joseph AI, Presley SH, Chaturvedi R, et al. The anti-inflammatory activity of curcumin is mediated by its oxidative metabolites. J Biol Chem. 2017;292:21243–52. doi: 10.1074/jbc.RA117.000123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Edwards RL, Luis PB, Nakashima F, Kunihiro AG, Presley S-H, Funk JL, et al. Mechanistic differences in the inhibition of NF-κB by turmeric and its curcuminoid constituents. J Agric Food Chem. 2020;68:6154–60. doi: 10.1021/acs.jafc.0c02607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Gordon ON, Luis PB, Ashley RE, Osheroff N, Schneider C. Oxidative transformation of demethoxy-and bisdemethoxycurcumin: products, mechanism of formation, and poisoning of human topoisomerase IIα. Chem Res Toxicol. 2015;28:989–96. doi: 10.1021/acs.chemrestox.5b00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Association AD. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes—2021. Diabetes Care. 2021;44:S15–33. doi: 10.2337/dc21-S002. [DOI] [PubMed] [Google Scholar]
  • 53. Maradana MR, Thomas R, O'Sullivan BJ. Targeted delivery of curcumin for treating type 2 diabetes. Mol Nutr Food Res. 2013;57:1550–6. doi: 10.1002/mnfr.201200791. [DOI] [PubMed] [Google Scholar]
  • 54. Lovshin JA, Drucker DJ. Incretin-based therapies for type 2 diabetes mellitus. Nat Rev Endocrinol. 2009;5:262–9. doi: 10.1038/nrendo.2009.48. [DOI] [PubMed] [Google Scholar]
  • 55. Fujiwara H, Hosokawa M, Zhou X, Fujimoto S, Fukuda K, Toyoda K, et al. Curcumin inhibits glucose production in isolated mice hepatocytes. Diabetes Res Clin Pract. 2008;80:185–91. doi: 10.1016/j.diabres.2007.12.004. [DOI] [PubMed] [Google Scholar]
  • 56. Sharma S, Kulkarni SK, Chopra K. Curcumin, the active principle of turmeric (Curcuma longa), ameliorates diabetic nephropathy in rats. Clin Exp Pharmacol Physiol. 2006;33:940–5. doi: 10.1111/j.1440-1681.2006.04468.x. [DOI] [PubMed] [Google Scholar]
  • 57. Takikawa M, Kurimoto Y, Tsuda T. Curcumin stimulates glucagon-like peptide-1 secretion in GLUTag cells via Ca2+/calmodulin-dependent kinase II activation. Biochem Biophys Res Commun. 2013;435:165–70. doi: 10.1016/j.bbrc.2013.04.092. [DOI] [PubMed] [Google Scholar]
  • 58. Tsuda T. Curcumin as a functional food-derived factor: degradation products, metabolites, bioactivity, and future perspectives. Food Funct. 2018;9:705–14. doi: 10.1039/c7fo01242j. [DOI] [PubMed] [Google Scholar]
  • 59. Chuengsamarn S, Rattanamongkolgul S, Luechapudiporn R, Phisalaphong C, Jirawatnotai S. Curcumin extract for prevention of type 2 diabetes. Diabetes Care. 2012;35:2121–7. doi: 10.2337/dc12-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res. 2011;30:1–14. doi: 10.1186/1756-9966-30-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Bauer JH, Helfand SL. New tricks of an old molecule: life-span regulation by p53. Aging Cell. 2006;5:437–40. doi: 10.1111/j.1474-9726.2006.00228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Tomeh MA, Hadianamrei R, Zhao X. A review of curcumin and its derivatives as anticancer agents. Int J Mol Sci. 2019;20:1033. doi: 10.3390/ijms20051033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Balasubramanian S, Eckert RL. Curcumin suppresses AP1 transcription factor-dependent differentiation and activates apoptosis in human epidermal keratinocytes. J Biol Chem. 2007;282:6707–15. doi: 10.1074/jbc.M606003200. [DOI] [PubMed] [Google Scholar]
  • 64. Jung EM, Lim JH, Lee TJ, Park J-W, Choi KS, Kwon TK. Curcumin sensitizes tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through reactive oxygen species-mediated upregulation of death receptor 5 (DR5) Carcinogenesis. 2005;26:1905–13. doi: 10.1093/carcin/bgi167. [DOI] [PubMed] [Google Scholar]
  • 65. Ravindran J, Prasad S, Aggarwal BB. Curcumin and cancer cells: how many ways can curry kill tumor cells selectively? AAPS J. 2009;11:495–510. doi: 10.1208/s12248-009-9128-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Labianca R, Beretta G, Gatta G, De Braud F, Wils J. Colon cancer. Crit Rev Oncol Hematol. 2004;51:145–70. doi: 10.1016/j.critrevonc.2004.03.003. [DOI] [PubMed] [Google Scholar]
  • 67. Selvam C, Prabu SL, Jordan BC, Purushothaman Y, Umamaheswari A, Zare MSH, et al. Molecular mechanisms of curcumin and its analogs in colon cancer prevention and treatment. Life Sci. 2019;239:117032. doi: 10.1016/j.lfs.2019.117032. [DOI] [PubMed] [Google Scholar]
  • 68. Carrato A. Adjuvant treatment of colorectal cancer. Gastrointest Cancer Res. 2008;2:S42. [PMC free article] [PubMed] [Google Scholar]
  • 69. Johnson JJ, Mukhtar H. Curcumin for chemoprevention of colon cancer. Cancer Lett. 2007;255:170–81. doi: 10.1016/j.canlet.2007.03.005. [DOI] [PubMed] [Google Scholar]
  • 70. Bäck M, Yin L, Ingelsson E. Cyclooxygenase-2 inhibitors and cardiovascular risk in a nation-wide cohort study after the withdrawal of rofecoxib. Eur Heart J. 2012;33:1928–33. doi: 10.1093/eurheartj/ehr421. [DOI] [PubMed] [Google Scholar]
  • 71. Lévesque LE, Brophy JM, Zhang B. The risk for myocardial infarction with cyclooxygenase-2 inhibitors: a population study of elderly adults. Ann Intern Med. 2005;142:481–9. doi: 10.7326/0003-4819-142-7-200504050-00113. [DOI] [PubMed] [Google Scholar]
  • 72. Garcea G, Berry DP, Jones DJ, Singh R, Dennison AR, Farmer PB, et al. Consumption of the putative chemopreventive agent curcumin by cancer patients: assessment of curcumin levels in the colorectum and their pharmacodynamic consequences. Cancer Epidemiol Biomark Prev. 2005;14:120–5. [PubMed] [Google Scholar]
  • 73. Sharma RA, Ireson CR, Verschoyle RD, Hill KA, Williams ML, Leuratti C, et al. Effects of dietary curcumin on glutathione S-transferase and malondialdehyde-DNA adducts in rat liver and colon mucosa: relationship with drug Levels1. Clin Cancer Res. 2001;7:1452–8. [PubMed] [Google Scholar]
  • 74. Ireson C, Orr S, Jones DJ, Verschoyle R, Lim C-K, Luo J-L, et al. Characterization of metabolites of the chemopreventive agent curcumin in human and rat hepatocytes and in the rat in vivo, and evaluation of their ability to inhibit phorbol ester-induced prostaglandin E2 production. Cancer Res. 2001;61:1058–64. [PubMed] [Google Scholar]
  • 75. Mudduluru G, George-William JN, Muppala S, Asangani IA, Kumarswamy R, Nelson LD, et al. Curcumin regulates miR-21 expression and inhibits invasion and metastasis in colorectal cancer. Biosci Rep. 2011;31:185–97. doi: 10.1042/BSR20100065. [DOI] [PubMed] [Google Scholar]
  • 76. Mosieniak G, Adamowicz M, Alster O, Jaskowiak H, Szczepankiewicz AA, Wilczynski GM, et al. Curcumin induces permanent growth arrest of human colon cancer cells: link between senescence and autophagy. Mech Ageing Dev. 2012;133:444–55. doi: 10.1016/j.mad.2012.05.004. [DOI] [PubMed] [Google Scholar]
  • 77. Bounaama A, Djerdjouri B, Laroche-Clary A, Le Morvan V, Robert J. Short curcumin treatment modulates oxidative stress, arginase activity, aberrant crypt foci, and TGF-β1 and HES-1 transcripts in 1, 2-dimethylhydrazine-colon carcinogenesis in mice. Toxicology. 2012;302:308–17. doi: 10.1016/j.tox.2012.08.014. [DOI] [PubMed] [Google Scholar]
  • 78. McFadden R-MT, Larmonier CB, Shehab KW, Midura-Kiela M, Ramalingam R, Harrison CA, et al. The role of curcumin in modulating colonic microbiota during colitis and colon cancer prevention. Inflamm Bowel Dis. 2015;21:2483–94. doi: 10.1097/MIB.0000000000000522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Cruz-Correa M, Hylind LM, Marrero JH, Zahurak ML, Murray-Stewart T, Casero RA, Jr, et al. Efficacy and safety of curcumin in treatment of intestinal adenomas in patients with familial adenomatous polyposis. Gastroenterology. 2018;155:668–73. doi: 10.1053/j.gastro.2018.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Carroll RE, Benya RV, Turgeon DK, Vareed S, Neuman M, Rodriguez L, et al. Phase IIa clinical trial of curcumin for the prevention of colorectal neoplasia. Cancer Prev Res. 2011;4:354–64. doi: 10.1158/1940-6207.CAPR-10-0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Kunnumakkara AB, Diagaradjane P, Guha S, Deorukhkar A, Shentu S, Aggarwal BB, et al. Curcumin sensitizes human colorectal cancer xenografts in nude mice to γ-radiation by targeting nuclear factor-κB–regulated gene products. Clin Cancer Res. 2008;14:2128–36. doi: 10.1158/1078-0432.CCR-07-4722. [DOI] [PubMed] [Google Scholar]
  • 82. Sandur SK, Deorukhkar A, Pandey MK, Pabón AM, Shentu S, Guha S, et al. Curcumin modulates the radiosensitivity of colorectal cancer cells by suppressing constitutive and inducible NF-κB activity. Int J Radiat Oncol Biol Phys. 2009;75:534–42. doi: 10.1016/j.ijrobp.2009.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Shakibaei M, Mobasheri A, Lueders C, Busch F, Shayan P, Goel A. Curcumin enhances the effect of chemotherapy against colorectal cancer cells by inhibition of NF-κB and Src protein kinase signaling pathways. PLoS One. 2013;8:e57218. doi: 10.1371/journal.pone.0057218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Shakibaei M, Buhrmann C, Kraehe P, Shayan P, Lueders C, Goel A. Curcumin chemosensitizes 5-fluorouracil resistant MMR-deficient human colon cancer cells in high density cultures. PLoS One. 2014;9:e85397. doi: 10.1371/journal.pone.0085397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Rawla P. Epidemiology of prostate cancer. World J Oncol. 2019;10:63. doi: 10.14740/wjon1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. H Lajis N, Abas F, Othman I, Naidu R. Mechanism of anticancer activity of curcumin on androgen-dependent and androgen-independent prostate cancer. Nutrients. 2020;12:679. doi: 10.3390/nu12030679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Dorai T, Cao YC, Dorai B, Buttyan R, Katz AE. Therapeutic potential of curcumin in human prostate cancer. III. Curcumin inhibits proliferation, induces apoptosis, and inhibits angiogenesis of LNCaP prostate cancer cells in vivo. Prostate. 2001;47:293–303. doi: 10.1002/pros.1074. [DOI] [PubMed] [Google Scholar]
  • 88. Mukhopadhyay A, Bueso-Ramos C, Chatterjee D, Pantazis P, Aggarwal BB. Curcumin downregulates cell survival mechanisms in human prostate cancer cell lines. Oncogene. 2001;20:7597–609. doi: 10.1038/sj.onc.1204997. [DOI] [PubMed] [Google Scholar]
  • 89. Ramachandran C, Fonseca HB, Jhabvala P, Escalon EA, Melnick SJ. Curcumin inhibits telomerase activity through human telomerase reverse transcritpase in MCF-7 breast cancer cell line. Cancer Lett. 2002;184:1–6. doi: 10.1016/s0304-3835(02)00192-1. [DOI] [PubMed] [Google Scholar]
  • 90. Liu Q, Loo WT, Sze S, Tong Y. Curcumin inhibits cell proliferation of MDA-MB-231 and BT-483 breast cancer cells mediated by down-regulation of NFκB, cyclinD and MMP-1 transcription. Phytomedicine. 2009;16:916–22. doi: 10.1016/j.phymed.2009.04.008. [DOI] [PubMed] [Google Scholar]
  • 91. Wang D, Veena MS, Stevenson K, Tang C, Ho B, Suh JD, et al. Liposome-encapsulated curcumin suppresses growth of head and neck squamous cell carcinoma in vitro and in xenografts through the inhibition of nuclear factor κB by an AKT-independent pathway. Clin Cancer Res. 2008;14:6228–36. doi: 10.1158/1078-0432.CCR-07-5177. [DOI] [PubMed] [Google Scholar]
  • 92. Wilken R, Veena MS, Wang MB, Srivatsan ES. Curcumin: a review of anti-cancer properties and therapeutic activity in head and neck squamous cell carcinoma. Mol Cancer. 2011;10:1–19. doi: 10.1186/1476-4598-10-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Li-duan Z, Qiang-song T, Cui-huan W. Growth inhibition and apoptosis inducing mechanisms of curcumin on human ovarian cancer cell line A2780. Chin J Integr Med. 2006;12:126–31. doi: 10.1007/BF02857359. [DOI] [PubMed] [Google Scholar]
  • 94. Yunos NM, Beale P, Yu JQ, Huq F. Synergism from sequenced combinations of curcumin and epigallocatechin-3-gallate with cisplatin in the killing of human ovarian cancer cells. Anticancer Res. 2011;31:1131–40. [PubMed] [Google Scholar]
  • 95. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J Neurosci. 2001;21:8370–7. doi: 10.1523/JNEUROSCI.21-21-08370.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. De R, Kundu P, Swarnakar S, Ramamurthy T, Chowdhury A, Nair GB, et al. Antimicrobial activity of curcumin against Helicobacter pylori isolates from India and during infections in mice. Antimicrob Agents Chemother. 2009;53:1592–7. doi: 10.1128/AAC.01242-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Zahedipour F, Hosseini SA, Sathyapalan T, Majeed M, Jamialahmadi T, Al-Rasadi K, et al. Potential effects of curcumin in the treatment of COVID-19 infection. Phytother Res. 2020;34:2911–20. doi: 10.1002/ptr.6738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Rocha FAC, de Assis MR. Curcumin as a potential treatment for COVID-19. Phytother Res. 2020;34:2085–7. doi: 10.1002/ptr.6745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Aggarwal BB, Surh Y-J, Shishodia S. The molecular targets and therapeutic uses of curcumin in health and disease. Springer Science & Business Media; 2007. [Google Scholar]
  • 100. Kurup VP, Barrios CS. Immunomodulatory effects of curcumin in allergy. Mol Nutr Food Res. 2008;52:1031–9. doi: 10.1002/mnfr.200700293. [DOI] [PubMed] [Google Scholar]
  • 101. Gupta SC, Patchva S, Koh W, Aggarwal BB. Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin Exp Pharmacol Physiol. 2012;39:283–99. doi: 10.1111/j.1440-1681.2011.05648.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Shehzad A, Khan S, Sup Lee Y. Curcumin molecular targets in obesity and obesity-related cancers. Future Oncol. 2012;8:179–90. doi: 10.2217/fon.11.145. [DOI] [PubMed] [Google Scholar]
  • 103. Kunati SR, Yang S, William BM, Xu Y. An LC–MS/MS method for simultaneous determination of curcumin, curcumin glucuronide and curcumin sulfate in a phase II clinical trial. J Pharm Biomed Anal. 2018;156:189–98. doi: 10.1016/j.jpba.2018.04.034. [DOI] [PubMed] [Google Scholar]
  • 104. Xu X-Y, Meng X, Li S, Gan R-Y, Li Y, Li H-B. Bioactivity, health benefits, and related molecular mechanisms of curcumin: current progress, challenges, and perspectives. Nutrients. 2018;10:1553. doi: 10.3390/nu10101553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Kocaadam B, Şanlier N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit Rev Food Sci Nutr. 2017;57:2889–95. doi: 10.1080/10408398.2015.1077195. [DOI] [PubMed] [Google Scholar]
  • 106. Additives EPoF, Food NSat. Scientific Opinion on the reevaluation of curcumin (E 100) as a food additive. EFSA J. 2010;8:1679. [Google Scholar]
  • 107. Program NT. NTP toxicology and carcinogenesis studies of turmeric oleoresin (CAS No. 8024-37-1)(major component 79%–85% curcumin, CAS No. 458-37-7) in F344/N rats and B6C3F1 mice (feed studies) Natl Toxicol Program Tech Rep Ser. 1993;427:1–275. [PubMed] [Google Scholar]
  • 108. Lao CD, Ruffin MT, Normolle D, Heath DD, Murray SI, Bailey JM, et al. Dose escalation of a curcuminoid formulation. BMC Complement Altern Med. 2006;6:1–4. doi: 10.1186/1472-6882-6-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Sharma RA, Euden SA, Platton SL, Cooke DN, Shafayat A, Hewitt HR, et al. Phase I clinical trial of oral curcumin: biomarkers of systemic activity and compliance. Clin Cancer Res. 2004;10:6847–54. doi: 10.1158/1078-0432.CCR-04-0744. [DOI] [PubMed] [Google Scholar]
  • 110. Adahoun MA, Al-Akhras MH, Jaafar MS, Bououdina M. Enhanced anti-cancer and antimicrobial activities of curcumin nanoparticles. Artif Cells Nanomed Biotechnol. 2017;45:98–107. doi: 10.3109/21691401.2015.1129628. [DOI] [PubMed] [Google Scholar]
  • 111. Joung HJ, Choi MJ, Kim JT, Park SH, Park HJ, Shin GH. Development of food-grade curcumin nanoemulsion and its potential application to food beverage system: antioxidant property and in vitro digestion. J Food Sci. 2016;81:N745–53. doi: 10.1111/1750-3841.13224. [DOI] [PubMed] [Google Scholar]
  • 112. Sari T, Mann B, Kumar R, Singh R, Sharma R, Bhardwaj M, et al. Preparation and characterization of nanoemulsion encapsulating curcumin. Food Hydrocoll. 2015;43:540–6. [Google Scholar]
  • 113. Thapa RK, Cazzador F, Grønlien KG, Tønnesen HH. Effect of curcumin and cosolvents on the micellization of Pluronic F127 in aqueous solution. Colloids Surf B Biointerfaces. 2020;195:111250. doi: 10.1016/j.colsurfb.2020.111250. [DOI] [PubMed] [Google Scholar]
  • 114. Bai F, Diao J, Wang Y, Sun S, Zhang H, Liu Y, et al. A new water-soluble nanomicelle formed through self-assembly of pectin–curcumin conjugates: preparation, characterization, and anticancer activity evaluation. J Agric Food Chem. 2017;65:6840–7. doi: 10.1021/acs.jafc.7b02250. [DOI] [PubMed] [Google Scholar]
  • 115. Lübtow MM, Nelke LC, Seifert J, Kühnemundt J, Sahay G, Dandekar G, et al. Drug induced micellization into ultra-high capacity and stable curcumin nanoformulations: physico-chemical characterization and evaluation in 2D and 3D in vitro models. J Control Release. 2019;303:162–80. doi: 10.1016/j.jconrel.2019.04.014. [DOI] [PubMed] [Google Scholar]
  • 116. Sehgal A, Kumar M, Jain M, Dhawan D. Modulatory effects of curcumin in conjunction with piperine on benzo (a) pyrene-mediated DNA adducts and biotransformation enzymes. Nutr Cancer. 2013;65:885–90. doi: 10.1080/01635581.2013.805421. [DOI] [PubMed] [Google Scholar]
  • 117. Hussain A, Somyajit K, Banik B, Banerjee S, Nagaraju G, Chakravarty AR. Enhancing the photocytotoxic potential of curcumin on terpyridyl lanthanide (III) complex formation. Dalton Trans. 2013;42:182–95. doi: 10.1039/c2dt32042h. [DOI] [PubMed] [Google Scholar]
  • 118. Valentini A, Conforti F, Crispini A, De Martino A, Condello R, Stellitano C, et al. Synthesis, oxidant properties, and antitumoral effects of a heteroleptic palladium (II) complex of curcumin on human prostate cancer cells. J Med Chem. 2009;52:484–91. doi: 10.1021/jm801276a. [DOI] [PubMed] [Google Scholar]
  • 119. Anand P, Thomas SG, Kunnumakkara AB, Sundaram C, Harikumar KB, Sung B, et al. Biological activities of curcumin and its analogues (Congeners) made by man and Mother Nature. Biochem Pharmacol. 2008;76:1590–611. doi: 10.1016/j.bcp.2008.08.008. [DOI] [PubMed] [Google Scholar]
  • 120. Yodkeeree S, Ampasavate C, Sung B, Aggarwal BB, Limtrakul P. Demethoxycurcumin suppresses migration and invasion of MDA-MB-231 human breast cancer cell line. Eur J Pharmacol. 2010;627:8–15. doi: 10.1016/j.ejphar.2009.09.052. [DOI] [PubMed] [Google Scholar]
  • 121. Luthra PM, Kumar R, Prakash A. Demethoxycurcumin induces Bcl-2 mediated G2/M arrest and apoptosis in human glioma U87 cells. Biochem Biophys Res Commun. 2009;384:420–5. doi: 10.1016/j.bbrc.2009.04.149. [DOI] [PubMed] [Google Scholar]
  • 122. Simstein R, Burow M, Parker A, Weldon C, Beckman B. Apoptosis, chemoresistance, and breast cancer: insights from the MCF-7 cell model system. Exp Biol Med. 2003;228:995–1003. doi: 10.1177/153537020322800903. [DOI] [PubMed] [Google Scholar]
  • 123. Lin C-C, Kuo C-L, Huang Y-P, Chen C-Y, Hsu M-J, Chu YL, et al. Demethoxycurcumin suppresses migration and invasion of human cervical cancer HeLa cells via inhibition of NF-κB pathways. Anticancer Res. 2018;38:2761–9. doi: 10.21873/anticanres.12519. [DOI] [PubMed] [Google Scholar]
  • 124. Chen Y, Hong C, Chen X, Qin Z. Demethoxycurcumin increases the sensitivity of cisplatin-resistant non-small lung cancer cells to cisplatin and induces apoptosis by activating the caspase signaling pathway. Oncol Lett. 2020;20:1. doi: 10.3892/ol.2020.12072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Kao CC, Cheng YC, Yang MH, Cha TL, Sun GH, Ho CT, et al. Demethoxycurcumin induces apoptosis in HER2 overexpressing bladder cancer cells through degradation of HER2 and inhibiting the PI3K/Akt pathway. Environ Toxicol. 2021;36:2186–95. doi: 10.1002/tox.23332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Hamzah H, Hertiani T, Pratiwi SUT, Murti YB, Nuryastuti T. The inhibition and degradation activity of demethoxycurcumin as antibiofilm on C. Albicans ATCC 10231. Res J Pharm Technol. 2020;13:377–82. [Google Scholar]
  • 127. Li Q-Q, Kang O-H, Kwon D-Y. Study on demethoxycurcumin as a promising approach to reverse methicillinresistance of Staphylococcus aureus. Int J Mol Sci. 2021;22:3778. doi: 10.3390/ijms22073778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Syu W-J, Shen C-C, Don M-J, Ou J-C, Lee G-H, Sun C-M. Cytotoxicity of curcuminoids and some novel compounds from Curcuma zedoaria. J Nat Prod. 1998;61:1531–4. doi: 10.1021/np980269k. [DOI] [PubMed] [Google Scholar]
  • 129. Devasena T, Rajasekaran K, Menon VP. Bis-1, 7-(2-hydroxyphenyl)-hepta-1, 6-diene-3, 5-dione (a curcumin analog) ameliorates DMH-induced hepatic oxidative stress during colon carcinogenesis. Pharmacol Res. 2002;46:39–45. doi: 10.1016/s1043-6618(02)00043-9. [DOI] [PubMed] [Google Scholar]
  • 130. Mahattanadul S, Nakamura T, Panichayupakaranant P, Phdoongsombut N, Tungsinmunkong K, Bouking P. Comparative antiulcer effect of bisdemethoxycurcumin and curcumin in a gastric ulcer model system. Phytomedicine. 2009;16:342–51. doi: 10.1016/j.phymed.2008.12.005. [DOI] [PubMed] [Google Scholar]
  • 131. Huang TY, Peng SF, Huang YP, Tsai CH, Tsai FJ, Huang CY, et al. Combinational treatment of all-trans retinoic acid (ATRA) and bisdemethoxycurcumin (BDMC)-induced apoptosis in liver cancer Hep3B cells. J Food Biochem. 2020;44:e13122. doi: 10.1111/jfbc.13122. [DOI] [PubMed] [Google Scholar]
  • 132. Li Y-B, Gao J-L, Zhong Z-F, Hoi P-M, Lee SM-Y, Wang Y-T. Bisdemethoxycurcumin suppresses MCF-7 cells proliferation by inducing ROS accumulation and modulating senescence-related pathways. Pharmacol Rep. 2013;65:700–9. doi: 10.1016/s1734-1140(13)71048-x. [DOI] [PubMed] [Google Scholar]
  • 133. Lai C-S, Chen Y-Y, Lee P-S, Kalyanam N, Ho C-T, Liou WS, et al. Bisdemethoxycurcumin inhibits adipogenesis in 3T3-L1 preadipocytes and suppresses obesity in high-fat diet-fed C57BL/6 mice. J Agric Food Chem. 2016;64:821–30. doi: 10.1021/acs.jafc.5b05577. [DOI] [PubMed] [Google Scholar]
  • 134. Wang S, Kang O-H, Kwon D-Y. Bisdemethoxycurcumin reduces methicillin-resistant Staphylococcus aureus expression of virulence-related exoproteins and inhibits the biofilm formation. Toxins (Basel) 2021;13:804. doi: 10.3390/toxins13110804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Lu PS, Inbaraj BS, Chen BH. Determination of oral bioavailability of curcuminoid dispersions and nano-emulsions prepared from Curcuma longa Linnaeus. J Sci Food Agric. 2018;98:51–63. doi: 10.1002/jsfa.8437. [DOI] [PubMed] [Google Scholar]
  • 136. Aggarwal BB, Deb L, Prasad S. Curcumin differs from tetrahydrocurcumin for molecular targets, signaling pathways and cellular responses. Molecules. 2014;20:185–205. doi: 10.3390/molecules20010185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Lai C-S, Ho C-T, Pan M-H. The cancer chemopreventive and therapeutic potential of tetrahydrocurcumin. Biomolecules. 2020;10:831. doi: 10.3390/biom10060831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Sugiyama Y, Kawakishi S, Osawa T. Involvement of the β-diketone moiety in the antioxidative mechanism of tetrahydrocurcumin. Biochem Pharmacol. 1996;52:519–25. doi: 10.1016/0006-2952(96)00302-4. [DOI] [PubMed] [Google Scholar]
  • 139. Okada K, Wangpoengtrakul C, Tanaka T, Toyokuni S, Uchida K, Osawa T. Curcumin and especially tetrahydrocurcumin ameliorate oxidative stress-induced renal injury in mice. J Nutr. 2001;131:2090–5. doi: 10.1093/jn/131.8.2090. [DOI] [PubMed] [Google Scholar]
  • 140. Naito M, Wu X, Nomura H, Kodama M, Kato Y, Kato Y, et al. The protective effects of tetrahydrocurcumin on oxidative stress in cholesterol-fed rabbits. J Atheroscler Thromb. 2002;9:243–50. doi: 10.5551/jat.9.243. [DOI] [PubMed] [Google Scholar]
  • 141. Nakmareong S, Kukongviriyapan U, Pakdeechote P, Donpunha W, Kukongviriyapan V, Kongyingyoes B, et al. Antioxidant and vascular protective effects of curcumin and tetrahydrocurcumin in rats with L-NAME-induced hypertension. Naunyn-Schmiedeberg’s Arch Pharmacol. 2011;383:519–29. doi: 10.1007/s00210-011-0624-z. [DOI] [PubMed] [Google Scholar]
  • 142. Chen J-W, Kong Z-L, Tsai M-L, Lo C-Y, Ho C-T, Lai C-S. Tetrahydrocurcumin ameliorates free fatty acid-induced hepatic steatosis and improves insulin resistance in HepG2 cells. J Food Drug Anal. 2018;26:1075–85. doi: 10.1016/j.jfda.2018.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Lai CS, Wu JC, Yu SF, Badmaev V, Nagabhushanam K, Ho CT, et al. Tetrahydrocurcumin is more effective than curcumin in preventing azoxymethane-induced colon carcinogenesis. Mol Nutr Food Res. 2011;55:1819–28. doi: 10.1002/mnfr.201100290. [DOI] [PubMed] [Google Scholar]
  • 144. Yoysungnoen P, Wirachwong P, Changtam C, Suksamrarn A, Patumraj S. Anti-cancer and anti-angiogenic effects of curcumin and tetrahydrocurcumin on implanted hepatocellular carcinoma in nude mice. World J Gastroenterol. 2008;14:2003–9. doi: 10.3748/wjg.14.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Masuda T, Hidaka K, Shinohara A, Maekawa T, Takeda Y, Yamaguchi H. Chemical studies on antioxidant mechanism of curcuminoid: analysis of radical reaction products from curcumin. J Agric Food Chem. 1999;47:71–7. doi: 10.1021/jf9805348. [DOI] [PubMed] [Google Scholar]
  • 146. Shen L, Ji H-F. The pharmacology of curcumin: is it the degradation products? Trends Mol Med. 2012;18:138–44. doi: 10.1016/j.molmed.2012.01.004. [DOI] [PubMed] [Google Scholar]
  • 147. Gordon ON, Schneider C. Vanillin and ferulic acid: not the major degradation products of curcumin. Trends Mol Med. 2012;18:361–3. doi: 10.1016/j.molmed.2012.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Alam MA, Sernia C, Brown L. Ferulic acid improves cardiovascular and kidney structure and function in hypertensive rats. J Cardiovasc Pharmacol. 2013;61:240–9. doi: 10.1097/FJC.0b013e31827cb600. [DOI] [PubMed] [Google Scholar]
  • 149. Bami E, Ozakpinar OB, Ozdemir-Kumral ZN, Köroglu K, Ercan F, Cirakli Z, et al. Protective effect of ferulic acid on cisplatin induced nephrotoxicity in rats. Environ Toxicol Pharmacol. 2017;54:105–11. doi: 10.1016/j.etap.2017.06.026. [DOI] [PubMed] [Google Scholar]
  • 150. Okuda M, Fujita Y, Sugimoto H. The additive effects of low dose intake of ferulic acid, phosphatidylserine and curcumin, not alone, improve cognitive function in APPswe/PS1dE9 transgenic mice. Biol Pharm Bull. 2019;42:1694–706. doi: 10.1248/bpb.b19-00332. [DOI] [PubMed] [Google Scholar]
  • 151. Salamanova E, Atanasova M, Dimitrov I, Doytchinova I. Effects of curcumin and ferulic acid on the folding of amyloid-β peptide. Molecules. 2021;26:2815. doi: 10.3390/molecules26092815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Meng G, Meng X, Ma X, Zhang G, Hu X, Jin A, et al. Application of ferulic acid for Alzheimer's disease: combination of text mining and experimental validation. Front Neuroinform. 2018;12:31. doi: 10.3389/fninf.2018.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Gao J, Yu H, Guo W, Kong Y, Li Q, Yang S, et al. The anticancer effects of ferulic acid is associated with induction of cell cycle arrest and autophagy in cervical cancer cells. Cancer Cell Int. 2018;18:1–9. doi: 10.1186/s12935-018-0595-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Burri J, Graf M, Lambelet P, Löliger J. Vanillin: more than a flavouring agent—a potent antioxidant. J Sci Food Agric. 1989;48:49–56. [Google Scholar]
  • 155. Arya SS, Rookes JE, Cahill DM, Lenka SK. Vanillin: a review on the therapeutic prospects of a popular flavouring molecule. Adv Trad Med. 2021;21:1–17. [Google Scholar]
  • 156. Ramadoss DP, Sivalingam N. Vanillin extracted from Proso and Barnyard millets induce apoptotic cell death in HT-29 human colon cancer cell line. NutrCancer. 2020;72:1422–37. doi: 10.1080/01635581.2019.1672763. [DOI] [PubMed] [Google Scholar]
  • 157. Naz H, Tarique M, Khan P, Luqman S, Ahamad S, Islam A, et al. Evidence of vanillin binding to CAMKIV explains the anti-cancer mechanism in human hepatic carcinoma and neuroblastoma cells. Mol Cell Biochem. 2018;438:35–45. doi: 10.1007/s11010-017-3111-0. [DOI] [PubMed] [Google Scholar]
  • 158. Chen H, Zheng J, Ma J. Vanillin ameliorates changes in HIF-1 α expression and neuronal apoptosis in a rat model of spinal cord injury. Restor Neurol Neurosci. 2019;37:21–9. doi: 10.3233/RNN-180879. [DOI] [PubMed] [Google Scholar]
  • 159. Lan X-B, Wang Q, Yang J-M, Ma L, Zhang W-J, Zheng P, et al. Neuroprotective effect of Vanillin on hypoxic-ischemic brain damage in neonatal rats. Biomed Pharmacother. 2019;118:109196. doi: 10.1016/j.biopha.2019.109196. [DOI] [PubMed] [Google Scholar]
  • 160. Tyagi AK, Prasad S, Majeed M, Aggarwal BB. Calebin A, a novel component of turmeric, suppresses NF-κB regulated cell survival and inflammatory gene products leading to inhibition of cell growth and chemosensitization. Phytomedicine. 2017;34:171–81. doi: 10.1016/j.phymed.2017.08.021. [DOI] [PubMed] [Google Scholar]
  • 161. Park S-Y, Kim DS. Discovery of natural products from Curcuma longa that protect cells from beta-amyloid insult: a drug discovery effort against Alzheimer's disease. J Nat Prod. 2002;65:1227–31. doi: 10.1021/np010039x. [DOI] [PubMed] [Google Scholar]
  • 162. Li Y, Li S, Han Y, Liu J, Zhang J, Li F, et al. Calebin-A induces apoptosis and modulates MAPK family activity in drug resistant human gastric cancer cells. Eur J Pharmacol. 2008;591:252–8. doi: 10.1016/j.ejphar.2008.06.065. [DOI] [PubMed] [Google Scholar]
  • 163. Buhrmann C, Popper B, Kunnumakkara AB, Aggarwal BB, Shakibaei M. Evidence that Calebin A, a component of curcuma longa suppresses NF-κB mediated proliferation, invasion and metastasis of human colorectal cancer induced by TNF-β (Lymphotoxin) Nutrients. 2019;11:2904. doi: 10.3390/nu11122904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Buhrmann C, Kunnumakkara AB, Popper B, Majeed M, Aggarwal BB, Shakibaei M. Calebin a potentiates the effect of 5-FU and TNF-β (Lymphotoxin α) against human colorectal cancer cells: potential role of NF-κB. Int J Mol Sci. 2020;21:2393. doi: 10.3390/ijms21072393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Reddy DN. Role of the normal gut microbiota. World J Gastroenterol. 2015;21:8787–803. doi: 10.3748/wjg.v21.i29.8787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Ferreira CM, Vieira AT, Vinolo MAR, Oliveira FA, Curi R, Martins FdS. The central role of the gut microbiota in chronic inflammatory diseases. J Immunol Res. 20142014 doi: 10.1155/2014/689492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Kennedy PJ, Cryan JF, Dinan TG, Clarke G. Irritable bowel syndrome: a microbiome-gut-brain axis disorder? World J Gastroenterol. 2014;20:14105. doi: 10.3748/wjg.v20.i39.14105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Larsen N, Vogensen FK, Van Den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5:e9085. doi: 10.1371/journal.pone.0009085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Everard A, Cani PD. Diabetes, obesity and gut microbiota. Best Pract Res Clin Gastroenterol. 2013;27:73–83. doi: 10.1016/j.bpg.2013.03.007. [DOI] [PubMed] [Google Scholar]
  • 170. Gurung M, Li Z, You H, Rodrigues R, Jump DB, Morgun A, et al. Role of gut microbiota in type 2 diabetes pathophysiology. EBioMedicine. 2020;51:102590. doi: 10.1016/j.ebiom.2019.11.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Pan M-H, Huang T-M, Lin J-K. Biotransformation of curcumin through reduction and glucuronidation in mice. Drug Metab Dispos. 1999;27:486–94. [PubMed] [Google Scholar]
  • 172. Shen L, Liu L, Ji H-F. Regulative effects of curcumin spice administration on gut microbiota and its pharmacological implications. Food Nutr Res. 2017;61:1361780. doi: 10.1080/16546628.2017.1361780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Zam W. Gut microbiota as a prospective therapeutic target for curcumin: a review of mutual influence. J Nutr Metab. 20182018 doi: 10.1155/2018/1367984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Feng W, Wang H, Zhang P, Gao C, Tao J, Ge Z, et al. Modulation of gut microbiota contributes to curcumin-mediated attenuation of hepatic steatosis in rats. Biochim Biophys Acta Gen Subj. 2017;1861:1801–12. doi: 10.1016/j.bbagen.2017.03.017. [DOI] [PubMed] [Google Scholar]
  • 175. An C-Y, Sun Z-Z, Shen L, Ji H-F. Biotransformation of food spice curcumin by gut bacterium Bacillus megaterium DCMB-002 and its pharmacological implications. Food Nutr Res. 2017;61:1412814. [Google Scholar]
  • 176. Ohno M, Nishida A, Sugitani Y, Nishino K, Inatomi O, Sugimoto M, et al. Nanoparticle curcumin ameliorates experimental colitis via modulation of gut microbiota and induction of regulatory T cells. PLoS One. 2017;12:e0185999. doi: 10.1371/journal.pone.0185999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Hong T, Zou J, Jiang X, Yang J, Cao Z, He Y, et al. Curcumin supplementation ameliorates bile cholesterol supersaturation in hamsters by modulating gut microbiota and cholesterol absorption. Nutrients. 2022;14:1828. doi: 10.3390/nu14091828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Lou Y, Zheng J, Hu H, Lee J, Zeng S. Application of ultra-performance liquid chromatography coupled with quad-rupole time-of-flight mass spectrometry to identify curcumin metabolites produced by human intestinal bacteria. J Chromatogr B. 2015;985:38–47. doi: 10.1016/j.jchromb.2015.01.014. [DOI] [PubMed] [Google Scholar]
  • 179. Hassaninasab A, Hashimoto Y, Tomita-Yokotani K, Kobayashi M. Discovery of the curcumin metabolic pathway involving a unique enzyme in an intestinal microorganism. Proc Natl Acad Sci USA. 2011;108:6615–20. doi: 10.1073/pnas.1016217108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Gao Y, Zhuang Z, Gao S, Li X, Zhang Z, Ye Z, et al. Tetrahydrocurcumin reduces oxidative stress-induced apoptosis via the mitochondrial apoptotic pathway by modulating autophagy in rats after traumatic brain injury. Am J Transl Res. 2017;9:887. [PMC free article] [PubMed] [Google Scholar]
  • 181. Zeng Y, Qiu F, Liu Y, Qu G, Yao X. Isolation and identification of phase 1 metabolites of demethoxycurcumin in rats. Drug Metab Dispos. 2007;35:1564–73. doi: 10.1124/dmd.107.015008. [DOI] [PubMed] [Google Scholar]
  • 182. Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, et al. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18:1606–14. doi: 10.1038/mt.2010.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Kalani A, Kamat PK, Kalani K, Tyagi N. Epigenetic impact of curcumin on stroke prevention. Metab Brain Dis. 2015;30:427–35. doi: 10.1007/s11011-014-9537-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Aqil F, Munagala R, Jeyabalan J, Agrawal AK, Gupta R. Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J. 2017;19:1691–702. doi: 10.1208/s12248-017-0154-9. [DOI] [PubMed] [Google Scholar]
  • 185. Whiteside TL. The potential of tumor-derived exosomes for noninvasive cancer monitoring: an update. Expert Rev Mol Diagn. 2018;18:1029–40. doi: 10.1080/14737159.2018.1544494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Kooijmans SA, Vader P, vanDommelen SM, van Solinge WW, Schiffelers RM. Exosome mimetics: a novel class of drug delivery systems. Int J Nanomedicine. 2012;7:1525–41. doi: 10.2147/IJN.S29661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Aqil F, Kausar H, Agrawal AK, Jeyabalan J, Kyakulaga A-H, Munagala R, et al. Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp Mol Pathol. 2016;101:12–21. doi: 10.1016/j.yexmp.2016.05.013. [DOI] [PubMed] [Google Scholar]
  • 188. Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol Ther. 2011;19:1769–79. doi: 10.1038/mt.2011.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Gupta A, Pulliam L. Exosomes as mediators of neuro-inflammation. J Neuroinflammation. 2014;11:1–10. doi: 10.1186/1742-2094-11-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Taverna S, Fontana S, Monteleone F, Pucci M, Saieva L, De Caro V, et al. Curcumin modulates chronic myelogenous leukemia exosomes composition and affects angiogenic phenotype via exosomal miR-21. Oncotarget. 2016;7:30420. doi: 10.18632/oncotarget.8483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Kalani A, Kamat P, Chaturvedi P, Tyagi S, Tyagi N. Curcumin-primed exosomes mitigate endothelial cell dysfunction during hyperhomocysteinemia. Life Sci. 2014;107:1–7. doi: 10.1016/j.lfs.2014.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Luan X, Sansanaphongpricha K, Myers I, Chen H, Yuan H, Sun D. Engineering exosomes as refined biological nano-platforms for drug delivery. Acta Pharmacol Sin. 2017;38:754–63. doi: 10.1038/aps.2017.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Oskouie MN, Aghili Moghaddam NS, Butler AE, Zamani P, Sahebkar A. Therapeutic use of curcumin-encapsulated and curcumin-primed exosomes. J Cell Physiol. 2019;234:8182–91. doi: 10.1002/jcp.27615. [DOI] [PubMed] [Google Scholar]
  • 194. Zhang H-G, Kim H, Liu C, Yu S, Wang J, Grizzle WE, et al. Curcumin reverses breast tumor exosomes mediated immune suppression of NK cell tumor cytotoxicity. Biochim Biophys Acta Mol Cell Res. 2007;1773:1116–23. doi: 10.1016/j.bbamcr.2007.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Liu C, Yan X, Zhang Y, Yang M, Ma Y, Zhang Y, et al. Oral administration of turmeric-derived exosome-like nano-vesicles with anti-inflammatory and pro-resolving bio-actions for murine colitis therapy. J Nanobiotechnol. 2022;20:1–17. doi: 10.1186/s12951-022-01421-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Gao C, Zhou Y, Chen Z, Li H, Xiao Y, Hao W, et al. Turmeric-derived nanovesicles as novel nanobiologics for targeted therapy of ulcerative colitis. Theranostics. 2022;12:5596. doi: 10.7150/thno.73650. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Food and Drug Analysis are provided here courtesy of Food and Drug Administration, Taiwan

RESOURCES