Table 5.
Biological Effects | Type of Study | Dose, Time, and Model | Main Biological Findings | Ref. |
---|---|---|---|---|
Antitumoral | In vitro | 10 and 20 µM; 24 h; human breast cancer MDA-MB-231 and Hs-578T cells | Attenuation of breast cancer-induced angiogenesis via inhibiting VEGF up-regulation of miR-124 reduces angiogenesis (inhibiting STAT3). | [140] |
In vitro | 5, 10, 20, and 40 µM; 24 h; MDA-MB-231 and BT-549 cells | C3G induces reversion of EMT characterized by phenotype modulation with increased epithelial marker E-ca and ZO-1 and decreased mesenchymal marker vimentin, N-ca, and EMT-associated transcription factors Snail1 and Snail2. | [141] | |
In vitro and in vivo | 5, 20, 50, 150, 300, and 500 µM; 12, 24, 48, and 72 h; human breast cancer cells, melanoma cells, human embryonic kidney 293 cells, mouse and human primary melanocytes, and human samples of melanoma | C3G treatment arrested the cell cycle at the G2/M phase by targeting cyclin B1 (CCNB1) and promoted apoptosis via ERβ in both mouse and human melanoma cell lines. | [142] | |
In vivo | 10, 20, 40, and 80 µM; Chinese hamster ovary cells, human colon cancer cell lines, human breast cancer cell lines, and human melanoma cell line | C3G binds to talin (a key regulator of integrins and cell adhesion) and promotes the interaction of talin with β1A-integrin. | [143] | |
In vitro | 10 and 40 μM; 24 h; MDA-MB-231 and MDA-MB-468 breast cancer cells | The EMT inhibition is related to the upregulation of KLF4, which has been reported to be an EMT suppressor in breast cancer cells. The upregulation of KLF4 expression by C3G involves transcriptional suppression of FBXO32. It was found that FBXO32 acted as a promoter of EMT and cell migration/invasion. | [144] | |
In vivo | Drosophila malignant RafGOFscrib −/− model. Purified C3G was added to standard food. Doses: C3G: 0.1 mg/mL or 0.4 mg/mL. |
Purified C3G inhibited tumor growth invasion, distant migration and prolongs the survival of tumor flies. C3G inhibited tumor invasion by reducing the MMP1 activity and through JNK pathway. | [145] | |
In vitro | HeLa cells by evaluating cell proliferation assay (C3G doses: 0–800 μg/mL) during 24, 48, and 72 h; apoptosis (Cy3G dose: 400 μg/mL), cell cycle, cell migration, and invasion evaluation (C3G dose: 400 μg/mL). | C3G combined with DPP induced apoptosis associated with the suppressed PI3K/AKT/mTOR signaling. DDP plus C3G treatment of HeLa cells can inhibit cell proliferation through cyclin D1 downregulation. Finally, this combined treatment could inhibit the migration and invasion associated with decreasing the protein of TIMP-1. |
[146] | |
In vivo | Induction of hepatic precancerous lesion (PCL) with diethylnitrosamine/2-acetylaminofluorene (DEN/2-AAF) in a Wistar rat model. C3G (not described source) at 10, 15, and 20 mg/kg/day. The measured parameters were alpha fetoprotein (AFP) levels and liver function biochemical analytes, and RNA panel differential expression was evaluated via qPCR. Histopathological examination of liver sections stained with H&E was also conducted. |
AFP levels were significantly decreased in the three C3G doses. Decreased ALT levels and increased serum albumin were found after C3G treatment. Moreover, C3G treatment decreases, in a dose-dependent manner, the mRNA expression of long non-coding RNA MALAT1 and tubulin gamma 1 and increases the miR-125b levels. Histologically, less discriminated dysplastic nodules were exhibited by liver sections of rats treated with C3G. |
[147] | |
In vitro | C3G or its metabolite protocatechuic acid (PCA) were tested at the following doses: 100, 200, and 400 μM in HepG2 cells. Cell viability after C3G and PCA treatments, LDH release, and apoptosis in HepG2 cells in which cytotoxicity was induced using 2-amino-3-methylimidazo [4,5-f]quinoline (IQ) were evaluated using CCK-8, LDH release, and flow cytometry assays, respectively. Tandem mass tag (TMT)-based proteomics was utilized to characterize the proteins and pathways associated with the improvement after C3G and PCA treatment. |
Exposure to IQ increased cytotoxicity and apoptosis in HepG2 cells, which were alleviated by C3G and PCA. C3G was more effective than PCA in protecting HepG2 cells against IQ-induced cytotoxicity and regulating the related signaling pathways. Proteomics and bioinformatics analyses and Western blot validation revealed that apoptosis-related signaling pathways played pivotal roles in the protective effect of C3G against the cytotoxicity of IQ. Moreover, XIAP was identified as a key target. XIAP acts as a potent apoptotic inhibitor by hampering the activation of caspases 3, 7, and 9. Molecular docking provided evidence that C3G affected the bindings of IQ and its carcinogenic metabolites to XIAP-BIR3 and contributed to the inhibition of apoptosis. |
[148] | |
In vitro | Cisplatin (DDP) dose: 5 μg/mL. C3G (>98% purity) dose: 400 μg/mL. Cervical cancer HeLa cells. Measurements of oxidative stress (CAT, SOD, and GSH-Px) and quantitation of gene expression of bax, bcl-2, Nrf2, and Keap1 genes were performed. |
C3G-DDP inhibited the activity of the antioxidant defense enzymes SOD, CAT, and GSH-Px. In parallel, C3G-DDP reduced GSH concentration while increasing the concentration of ROS and MDA. C3G-DDP reduces the expression of Nrf2 and Nrf2 target proteins: HO-1 and NQO1. Finally, C3G-DDP increased the mRNA expression ratio of bax/bcl-2 and activated the intrinsic apoptotic pathway of HeLa cells. |
[149] | |
Antidiabetic and protection against complications of diabetes | In vitro and in vivo | 1 and 5 µM; 4 h pre-treatment of ARPE-19 cells exposed to 30 µM 4-hydroxynonenal for 24 h. 50 (mg/kg)/day for 3 weeks (2 pre-illumination and 1 post-illumination) in rabbits in which retinal damage was induced by light exposure | Decreased apoptosis, lower senescence-associated beta-galactosidase, and lower VEGF release. Increased thickness of the neurosensory retina in rabbits exposed to light. |
[150] |
In vitro | 10 µM; 6 h; mouse colonic epithelial MCE301 cells | Higher gene expression of the Mg2+ transport carriers Trpm6 and Cnnm4. | [151] | |
In vivo | 10 and 20 (mg/kg)/day for 8 weeks in Sprague Dawley rats in which diabetes was induced with a 45 mg/kg streptozotocin dose. | Reduced fasting glycemia and insulin levels, decreased serum creatinine and BUN, and lower urinary albumin. Improved antioxidant enzyme and reduced cytokine levels. Decreased fibrosis and glomerulosclerosis in renal tissue. | [152] | |
In vitro | 100 µM; 24 h; human corneal epithelial cells (HCEC 6510) previously exposed to 10 µg/mL of LPS for 24 h | Reduced apoptosis and decreased production of cytokines. | [153] | |
In vivo | 1.6 mg/mL in drinking water (∼6.4 mg/day), for 3 or 20 weeks, in C57BL/6J male mice fed a low- or high-fat diet | Decreased weight gain for high-fat diet, improved glucose tolerance, reduced hepatic and plasma triglycerides, and modulated hepatic FGF21 levels. | [154] | |
In vitro and in vivo | 20 µM; 24 h; HUVEC cells exposed for 1 h to 100 ng/mL TNF-alpha before treatment with C3G. 50 or 100 mg/kg; 8 weeks; male New Zealand rabbits fed 8 weeks with a high-fat diet after balloon catheter injury was performed. |
Reduced damage in the intima media; decreased levels of circulating cholesterol, low-density lipoprotein, and triglycerides; and increased high-density lipoprotein. Reduced levels of cytokines and lowered apoptosis rates. Higher expression of SIRT1. | [155] | |
In vitro | 20 µM; 48 h (+1 h pre-treatment); lens epithelial SRA01/04 cells exposed to 100 mM glucose and Sprague Dawley rat lens tissue exposed to 50 mM glucose. | Reduced apoptosis rates, decreased NFkB levels, and lowered Cox-2 protein expression. Decreased opacity of rat’s lens tissue. | [156] | |
In vitro | 5 and 10 µM pre-treatment; 24 h; 3T3-L1 cells and human SGBS cells exposed to 1 mM or 500 µM palmitate for 24 h | Reduced lipid content, lower PPARgamma and nuclear NFkB protein levels, improved levels of insulin signaling targets, and higher Adipoq gene expression. | [157] | |
In vivo | 10 and 50 µM; 24 h; HepG2 pre-treated with 400 µM palmitic acid and 400 µM oleic acid for 24 h. 50 mg/day; 8 weeks; male C57BL 6J mice previously fed an HFD for 4 weeks and 8 additional weeks of HFD during C3G treatment. | Reduced plasma and liver triglycerides, reduced fatty acid synthesis, lower fasting plasma glucose and insulin, higher cell glucose uptake, activation of PPAR-alpha. | [158] | |
Liver disease and hepato-protection | In vitro and in vivo | 100 µM; 12 h; HepG2 or AML-12 cells co-treated with 400 µM palmitic acid 0.2% (v/v) of C3G in the HFD, 4 weeks (after 12 weeks of HFD), male mice fed an HFD for 16 weeks | Reduced liver steatosis, lower fasting glucose and insulin levels, reduced NLRP3 inflammasome, higher antioxidative enzyme levels, lower ROS levels, increased mitophagy. | [159] |
In vitro | 5 µg/mL; 12 h; HepG2 cells pre-exposed to 4 µM hydrogen peroxide for 6 h | Decreased ROS levels, increased glutathione content, and higher catalase activity. Increased Nrf2 and Keap1 protein levels. | [160] | |
In vitro | 2.5–10 µM; 24 h; HepG2 cells previously treated with 400 µM hydrogen peroxide | Increased cell viability and antioxidative machinery. Decreased ROS, apoptosis rates, and apoptosis-related proteins. | [161] | |
In vitro and in vivo | 200 (mg/kg)/day; 8 weeks; male C57BL 6J mice fed an HFC diet and 5% ethanol drinking solution during the C3G treatment. HepG2 and FL83B cells were treated with SIRT1 inhibitor EX527 at 10 µM, for 4 h, and 1 µM C3G for an additional 20 h. |
Reduced liver lipid content, lower levels of proinflammatory cytokines and inflammasome proteins, reduced NFkB protein expression and acetylation, and increased SIRT1 protein levels. | [162] | |
Colitis and gastrointestinal alterations | In vivo and in vitro | 1 ug i.p. on days 0, 3, and 6 of model induction; C57BL 6J mice in which colitis was induced with drinking water containing 3.5% of dextran sulfate sodium for 7 days. 1 µg/mL; 24 h; peritoneal macrophages activated with 1 ug mL−1 of LPS. | Reduced cytokine gene expression in the colon, induction of Treg cells, and reduction of peritoneal CD169+ macrophages. | [163] |
In vivo | 500 and 1000 mg/kg of diet; 8 weeks; male Wistar rats in which dysbiosis and intestinal damage were parallelly induced with 20 mg/kg 3-chloro-1,2-propanediol for 8 weeks | Improved histological features, modulation of gut microbiota. | [164] | |
In vivo and in vitro | 50, 100, or 200 µmol/kg; 3 days; female BALB c mice in which colitis was induced with 2.5 mg of 2,4,6-trinitrobenzen-osulfonic acid 12 h after the first dose of C3G. 50 and 100 µmol/L; 24 h pre-treatment; LPS-induced Caco-2 cells with 100 ng/mL for 24 h. | Prevention of histological damage, reduction of proinflammatory cytokines, and suppression of nitric oxide production. | [165] | |
In vitro | 10 or 20 µmol/L; 24 h pre-treatment; Caco-2 cells induced with palmitic acid 100 µmol/L for 6 h | Decreased nuclear NFkB, reduction of cytokine IL6 and IL8 gene expression and COX2 protein, decrease in ROS, and increase in Nrf2 levels. | [166] | |
Neuroprotective | In vitro | 2.5, 5, or 10 µmol/L; 4 h pre-treatment; microglial BV2 (macrophage) cells stimulated with 1 μg/mL LPS for 24–48 h | Decreased cytokine levels, reduced iNOS mRNA levels and lower NO production, suppression of NFkB activation and p38 signaling pathway, decreased neurotoxicity and apoptosis in PC12 cells exposed to conditioned media from LPS-activated BV2 cells. | [167] |
In vitro | 0.05, 0.1, 0.25, 0.5, or 1 µmol/L; 24 h pre-treatment; HT22 neuronal cells exposed to 5 mM glutamate for 18 h | Reduction of apoptosis, decrease in ROS, increase in Nrf2 levels and antioxidative gene expression, reduction of ER stress biomarkers. | [168] | |
In vitro | 1, 3, or 9 µmol/L; 24–48 h; PC12 neuronal cells exposed in parallel to amyloid beta fibrils | Increased cell viability, decreased necrosis, reduced ROS levels. | [169] | |
In vitro | 30 mg kg-1 day-1; 38 weeks; APPswe/PS1dE9 mice modeling Alzheimer’s disease | Differential gene expression in the spleen of the treated animals, including upregulation of antioxidant and immune system-related molecular targets. | [170] | |
Reproductive health | In vitro | 5, 20, 40, 80, or 160 µmg/L; 2 h pre-treatment; Leydig R2C cells exposed to 44.8 µmol/L cadmium sulfate for 24 h | Increased cell viability, reduced ROS levels, protection of mitochondrial potential, increased StAR protein and progesterone levels. | [171] |
In vivo | 500 mg/kg of chow diet; 10, 20, or 30 days; Kunming male mice treated with 5 (mg/kg)/day of cadmium chloride | Decreased levels of circulating FSH and testosterone, increased LH circulating levels, differential modulation of gene expression in the hypothalamus, increased expression of proteins involved in testosterone biosynthesis. | [172] | |
Respiratory system, antiviral, and anti-SARS-CoV2 | In vivo | Diet containing 0.4% C3G (~1.2 mg/day); 25 days; asthma model of BALB/c mice sensitized to ovalbumin intraperitoneally (20 μg on days 0, 7, and 14) and nasally (1% aerosols on days 21–25) | Decreased number of peripheral eosinophils; reduced inflammatory infiltration in the lungs; lower levels of IL-4, IL-5, and IL-13; inhibition of IL-4Ra-STAT6 pathway. | [173] |
In silico and in vitro | 3–200 µmol/L; papain-like protease assay for determination of deubiquitinase activity | The molecular docking prediction showed a potential binding activity to the papain-like protease of SARS-CoV-2, concentration-dependent inhibition of papain-like protease deubiquitinase activity. | [174] | |
In silico and in vitro | 3–200 µmol/L; papain-like protease assay for determination of total protease activity | Molecular docking prediction of binding to the papain-like protease of SARS-CoV2. Concentration-dependent inhibition of papain-like protease total protease activity. | [175] | |
In vivo and in vitro | 200 or 400 mg/kg bw; oral administration from days 2–28; Sprague Dawley male rats injected intraperitoneally with monocrotaline 60 mg/kg bw on day 1 to induce a model of pulmonary artery hypertension. 10 or 20 µmol/L; 24 h pre-treatment; cells induced with TGF-beta1 8 ng/mL for additional 24 h. |
Reduction of hemodynamic indicators of pulmonary artery hypertension, improved histological features and blood oxygenation, reduction of cytokines levels, reduced markers of proliferation in PASMC, inhibition of TGF-beta1-p38 MAPK-CREB signaling pathway. | [176] | |
Anti-inflammatory and immune system modulation | In vivo and in vitro | 25 mg/kg; two tail-vein injections per week for a total of six injections starting ten days after the secondary immunization; Sprague-Dawley male rats in which arthritis was induced by three injections of bovine type II collagen. 25, 50, or 100 µmol/L; 24–48 h; rheumatoid arthritis synovial fibroblasts and mononuclear cells obtained from patients. |
Increased Treg cells and decreased CD38+ NK cell proportion in blood and synovial fluid in murine model, increased apoptosis and decreased proliferation in human rheumatoid arthritis synovial fibroblasts, decrease in proinflammatory cytokines. | [177] |
In vivo | 10 (mg/kg)/day; 15 weeks; spontaneously hypertensive male rats and Wistar-Kyoto rats. | No differences were observed either in the spleen weights or in the proportions of splenic T-cells and helper T-cells; modulation of CD62Lhi, CD62Llo, CD62L-, CD25+, and T-reg cells dependent on the genotype. | [178] | |
In vitro and in vivo | 25, 50, 100, and 250 µmol/L, RBL-2H3 cells sensitized with anti-DNP IgE and exposed to DNP-BSA antigen 100 and 200 µmol/kg bw, orally administered 1 h before antigen exposure, and 40 mg/kg bw, intravenous administration 1 h before antigen challenge, male ICR mice sensitized with anti-DNP IgE (100 ng injection in the ear) 24 h before the experiment and then challenged with DNP-BSA antigen (140 µg/mouse). | Dose-dependent inhibition of histamine and beta-hexosaminidase release, decreased ear tissue response (measured as extravasation) after antigen challenge. | [179] | |
Other studies | In vitro | 80 µmol/L; 2 h pre-treatment; primary human dermal fibroblast irradiated with 12 J/cm2 UVA light and treated with 3-methyladenine | Decreased apoptosis, increased expression of autophagy markers, reduced ROS levels. | [180] |
In vivo | 100 mg/kg body weight, oral administration before induction; Wistar rats injected with 1 mL/kg of 5% taurocholate to induce a model of severe acute pancreatitis | Increased colonic motility, decreased serum levels of H2S and pro-inflammatory cytokines, activation of mTOR signaling, reduced protein levels of cystathionine-gamma-lyase. | [181] | |
In silico | Molecular modeling to assess for potential interactions between C3G and the advanced glycation end product receptor and its ligands | The results suggest a potential interaction and subsequent inhibition of the receptor for advanced glycation end products. | [182] | |
In vitro | 25–400 µM; 24–72 h; primary human osteoblasts and MC3T3-E1 osteoblast murine cell line | Increased cell proliferation, increased mineralization activity, activation of ERK1/2 signaling pathway, increased osteocalcin protein and mRNA levels. | [183] | |
In vitro | 1.25, 2.5, and 5 µmol/L; 24 h co-treatment or 2 h pre-treatment; primary human articular chondrocytes exposed to advanced glycation end products 10 µg/mL for 24 h (parallel to C3G treatment) or 10 min (after 2 h pre-treatment). | Reduced protein and mRNA expression levels of matrix metalloproteinases, decreased NF-kB signaling, reduced ERK/MAPK signaling activation. | [184] | |
In vitro | 20 µmol/L; six-day treatment renewed every 48 h; human amniotic epithelial cells | Differential modulation of genes including targets involved in adipocyte differentiation and muscle activity. | [185] |
EMT = epithelial–mesenchymal transition, CCNB1 = cyclin B1, DDP = cis-diamminedichloroplatinum, CAT = catalase, SOD = superoxide dismutase, GSH-Px = glutathione peroxidase, Nrf2 = nuclear factor erythroid 2-related factor-2, Keap1 = Kelch-like ECH-associated protein 1, HO-1 = heme oxygenase-1, NOQ1 = NAD(P)H quinone dehydrogenase 1, PCA = protocatechuic acid, MDA = malondialdehyde, TMT = tandem mass tag, VEGF = vascular endothelial growth factor.