Phytotherapy and the Role of Bioactive Compounds in Modulating Mechanisms of Overweight and Obesity Comorbid with Depressive Symptoms—A Scoping Review of Mechanisms of Action
Abstract
1. Introduction
2. Materials and Methods
3. Epidemiological and Public Health Impact of Obesity and Depression
4. The Role of Major Classes of Bioactive Compounds in the Treatment of Obesity and Depression
5. The Role of Bioactive Compounds in the Management of Obesity and Depression
The Role and Classification of Bioactive Compounds in Therapy
6. Selected Bioactive Compounds and Their Mechanisms of Action
6.1. The Treatment of Obesity and Its Mechanisms of Action
6.2. Treatments for Depression Comorbid with Obesity and Their Mechanisms of Action
Compound | Natural Source | Mechanism of Action | References |
---|---|---|---|
Quercetin | Onions, apples, and berries | Antioxidant, anti-inflammatory, modulates AMPK and cytokines | [67,68,69,70,71,72,73,74,75,76,77,78,79,80] |
Apigenin | Parsley, celery, and chamomile | Anxiolytic, neuroprotective, regulates BDNF, mTOR/AMPK/ULK1 | [133,134,135,136,137] |
Luteolin | Celery, green pepper, and chamomile | Reduces neuroinflammation, affects ER stress, and monoamine transporters | [159,160,161,162,163,164] |
Naringenin | Citrus fruits | Antioxidant, modulates BDNF signaling, antidepressant | [138,139,140,141,142,143,144] |
Baicalin | Scutellaria baicalensis | Anti-inflammatory, modulates PI3K/Akt, CREB/BDNF, and BBB protection | [165,166,167,168,169,170] |
Genistein | Soy products, soybeans | Phytoestrogenic, affects neurotransmitter systems, regulates lipid metabolism | [81,82,83,84,85,86,87,88,89] |
Resveratrol | Grapes, red wine, berries | Activates SIRT1 and AMPK, anti-adipogenic, neuroprotective | [90,91,92,93,94,95,96,97,98,99,100,101] |
Curcumin | Turmeric | Anti-inflammatory, modulates NF-κB, serotonin, and dopamine | [145,146,147,148,149,150,151,152] |
EGCG | Green tea | Modulates metabolism, anti-obesity, and neuroprotective, insulin sensitivity | [102,103,104,105,106,107] |
Anthocyanins | Berries, red cabbage, purple corn | Anti-inflammatory, modulate adipogenesis and lipid metabolism, antioxidant, influence gut-brain axis | [102,103,104,105,106,107] |
Betalains | Beetroot, pitaya | Antioxidant, anti-inflammatory, inhibits peroxynitrite | [118,119,120,121,122,123,124] |
Piperine | Black pepper | Enhances bioavailability, MAO inhibition, neuroactive | [153,154,155,156,157,158] |
Omega-3 fatty acids | Fish oil, flaxseed | Anti-inflammatory, neuroprotective | [170,171,172,173,174,175,176,177,178] |
7. In Vitro and In Vivo Studies on Bioactive Compounds
7.1. In Vitro Studies
7.2. In Vivo Studies
8. Personalized Diets Enriched with Bioactive Compounds and Probiotics in the Management of Obesity and Depression
9. Gut Microbiota Modulation by Bioactive Compounds and Probiotics
10. The Gut–Brain Axis and Translational Potential Perspectives
Synbiotic-Based Strategies and Clinical Implementation
11. Functional Diets as a Therapeutic Strategy
Toward Clinical Applications and Functional Product Development
- Phase I/II studies assessing the safety, tolerability, and preliminary efficacy of combined nutraceuticals (e.g., EGCG + curcumin + omega-3 fatty acids) in obese individuals with depressive symptoms;
- Randomized controlled trials (RCTs) evaluating synbiotic approaches (e.g., anthocyanins or flavonoids + Lactobacillus rhamnosus or Bifidobacterium breve) targeting inflammation and the gut–brain axis;
- Comparative studies testing phytochemical-enriched diets versus standard pharmacotherapy in patients with mild-to-moderate depression and insulin resistance.
12. Pharmacokinetics and the Potential Side Effects of Selected Phyto-Chemicals: Potential Adverse Effects of Major Compounds (Curcumin, EGCG, and Resveratrol)
12.1. Adverse Effects of Resveratrol
12.2. Adverse Effects of Epigallocatechin Gallate
12.3. Adverse Effects of Curcumin
13. Limitations
14. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Chan, M. Obesity and Diabetes: The Slow-Motion Disaster. Milbank Q. 2017, 95, 11–14. [Google Scholar] [CrossRef] [PubMed]
- Schwarzenberg, S.J.; Sinaiko, A.R. Obesity and inflammation in children. Paediatr. Respir. Rev. 2006, 7, 239–246. [Google Scholar] [CrossRef]
- Jantaratnotai, N.; Mosikanon, K.; Lee, Y.; McIntyre, R.S. The interface of depression and obesity. Obes. Res. Clin. Pract. 2017, 11, 1–10. [Google Scholar] [CrossRef]
- Luppino, F.S.; de Wit, L.M.; Bouvy, P.F.; Stijnen, T.; Cuijpers, P.; Penninx, B.W.J.H.; Zitman, F.G. Overweight, obesity, and depression: A systematic review and meta-analysis of longitudinal studies. Arch. Gen. Psychiatry 2010, 67, 220–229. [Google Scholar] [CrossRef]
- Joseph, S.V.; Edirisinghe, I.; Burton-Freeman, B.M. Fruit Polyphenols: A Review of Anti-inflammatory Effects in Humans. Crit. Rev. Food Sci. Nutr. 2016, 56, 419–444. [Google Scholar] [CrossRef] [PubMed]
- Shelton, R.C.; Miller, A.H. Inflammation in depression: Is adiposity a cause? Dialogues Clin. Neurosci. 2011, 13, 41–53. [Google Scholar] [CrossRef]
- Noori, T.; Sureda, A.; Sobarzo-Sánchez, E.; Shirooie, S. The Role of Natural Products in Treatment of Depressive Disorder. Curr. Neuropharmacol. 2022, 20, 929–949. [Google Scholar] [CrossRef]
- Lim, S.S.; Vos, T.; Flaxman, A.D.; Danaei, G.; Shibuya, K.; Adair-Rohani, H.; AlMazroa, M.A.; Amann, M.; Anderson, H.R.; Andrews, K.G.; et al. Global and regional burden of diseases and injuries in 2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2095–2128. [Google Scholar] [CrossRef]
- Olszanecka-Glinianowicz, M. Depression—Cause or result of obesity? Endokrynol. Otyłość Zab. Przem. Mat. 2008, 4, 78–85. [Google Scholar]
- Ekinci, G.N.; Sanlier, N. The relationship between nutrition and depression in the life process: A mini-review. Exp. Gerontol. 2023, 172, 112072. [Google Scholar] [CrossRef]
- Kazimierska, I. Otyłość to choroba, a nie defekt estetyczny. Kurier Medyczny 2021, 2, 14–15. [Google Scholar]
- Kopacz, M.S.; Wichowicz, H.M. Obesity—Causes, Types, Treatment, Effects. Eng. Sci. Technol. 2018, 4, 65–79. [Google Scholar] [CrossRef]
- Stunkard, A.J.; Faith, M.S.; Allison, K.C. Depression and obesity. Biol. Psychiatry 2003, 54, 330–337. [Google Scholar] [CrossRef] [PubMed]
- Mayorga, P. El Adipocito Como Órgano Endocrino: Implicaciones Fisiopatológicas y Terapéuticas. Rev. Med. 2007, 152, 225–242. [Google Scholar]
- Stolarczyk, E. Adipose tissue inflammation in obesity: A metabolic or immune response? Curr. Opin. Pharmacol. 2017, 37, 35–40. [Google Scholar] [CrossRef]
- Ghasemzadeh, A.A. Flavonoids and phenolic acids: Role and biochemical activity in plants and humans. J. Med. Plants Res. 2011, 5, 6697–6703. [Google Scholar] [CrossRef]
- Stalikas, C.D. Extraction, separation, and detection methods for phenolic acids and flavonoids. J. Sep. Sci. 2007, 30, 3268–3295. [Google Scholar] [CrossRef]
- Tanase, C.; Cosarcă, S.; Muntean, D.L. A critical review of phenolic compounds extracted from the shoulder of woody vascular plants and their potential biological activity. Molecules 2019, 24, 1182. [Google Scholar] [CrossRef] [PubMed]
- Aranaz, P.; Navarro-Herrera, D.; Zabala, M.; Miguéliz, I.; Romo-Hualde, A.; López-Yoldi, M.; Alfredo Martínez, J.; Vizmanos, J.L.; Milagro, F.I.; González-Navarro, C.J. Phenolic compounds inhibit 3T3-L1 adipogenesis depending on the stage of differentiation and their binding affinity to PPARγ. Molecules 2019, 24, 1045. [Google Scholar] [CrossRef]
- Hsu, C.L.; Yen, G.C. Effects of flavonoids and phenolic acids on the inhibition of adipogenesis in 3T3-L1 adipocytes. J. Agric. Food Chem. 2007, 55, 8404–8410. [Google Scholar] [CrossRef]
- Alam, M.A.; Subhan, N.; Hossain, H.; Hossain, M.; Reza, H.M.; Rahman, M.M.; Ullah, M.O. Hydroxycinnamic acid derivatives: A potential class of natural compounds for the management of lipid metabolism and obesity. Nutr. Metab. 2016, 13, 27. [Google Scholar] [CrossRef]
- Ibitoye, O.B.; Ajiboye, T.O. Dietary phenolic acids reverse insulin resistance, hyperglycaemia, dyslipidaemia, inflammation and oxidative stress in high-fructose diet-induced metabolic syndrome rats. Arch. Physiol. Biochem. 2018, 124, 410–417. [Google Scholar] [CrossRef]
- Lee, W.; Lee, S.Y.; Son, Y.-J.; Yun, J.-M. Gallic acid decreases inflammatory cytokine secretion through histone acetyltransferase/histone deacetylase regulation in high glucose-induced human monocytes. J. Med. Food 2015, 18, 793–801. [Google Scholar] [CrossRef]
- Saura-Calixto, F. Antioxidant dietary fiber product: A new concept and potential food ingredient. J. Agric. Food Chem. 1998, 46, 4303–4306. [Google Scholar] [CrossRef]
- Eskicioglu, V.; Kamiloglu, S.; Nilufer-Erdil, D. Antioxidant dietary fibres: Potential functional food ingredients from plant processing by-products. Czech J. Food Sci. 2016, 33, 487–499. [Google Scholar] [CrossRef]
- Ozyurt, V.; Ye, H.; Ötles, S. Effect of food processing on the physicochemical properties of dietary fibre. Acta Sci. Pol. Technol. Aliment. 2016, 15, 233–245. [Google Scholar] [CrossRef]
- Requena, M.C.; González, C.N.A.; Barragán, L.A.P.; Correia, T.; Esquivel, J.C.C.; Herrera, R.R. Functional and physico-chemical properties of six desert-sources of dietary fiber. Food Biosci. 2016, 16, 26–31. [Google Scholar] [CrossRef]
- Zhou, Q.; Wu, J.; Tang, J.; Wang, J.-J.; Lu, C.-H.; Wang, P.-X. Beneficial effect of higher dietary fiber intake on plasma HDL-C and TC/HDL-C ratio among Chinese rural-to-urban migrant workers. Int. J. Environ. Res. Public Health 2015, 12, 4726. [Google Scholar] [CrossRef]
- Adam, C.L.; Thomson, L.M.; Williams, P.A.; Ross, A.W. Soluble Fermentable Dietary Fibre (Pectin) Decreases Caloric Intake, Adiposity and Lipidaemia in High-Fat Diet-Induced Obese Rats. PLoS ONE 2015, 10, e0140392. [Google Scholar] [CrossRef]
- Aoe, S.; Ichinose, Y.; Kohyama, N.; Komae, K.; Takahashi, A.; Abe, D.; Yoshioka, T.; Yanagisawa, T. Effects of high β-glucan barley on visceral fat obesity in Japanese individuals: A randomized, double-blind study. Nutrition 2017, 42, 1–6. [Google Scholar] [CrossRef]
- Im, H.J.; Yoon, K.Y. Production and characterisation of alcohol-insoluble dietary fibre as a potential source for functional carbohydrates produced by enzymatic depolymerisation of buckwheat hulls. Czech J. Food Sci. 2016, 33, 449–457. [Google Scholar] [CrossRef]
- Mosikanon, K.; Arthan, D.; Kettawan, A.; Tungtrongchitr, R.; Prangthip, P. Yeast β-glucan modulates inflammation and waist circumference in overweight and obese subjects. J. Diet. Suppl. 2017, 14, 173–185. [Google Scholar] [CrossRef] [PubMed]
- Samout, N.; Ettaya, A.; Bouzenna, H.; Ncib, S.; Elfeki, A.; Hfaiedh, N. Beneficial effects of Plantago albicans on high-fat diet-induced obesity in rats. Biomed. Pharmacother. 2016, 84, 1768–1775. [Google Scholar] [CrossRef] [PubMed]
- Zou, J.; Chassaing, B.; Singh, V.; Pellizzon, M.; Ricci, M.; Fythe, M.D.; Kumar, M.V.; Gewirtz, A.T. Fiber-mediated nutrition of the gut microbiota protects against diet-induced obesity by restoring IL-22-dependent colonic health. Cell Host Microbe 2018, 23, 41–53.e4. [Google Scholar] [CrossRef]
- Sánchez, D.; Quiñones, M.; Moulay, L.; Muguerza, B.; Miguel, M.; Aleixandre, A. Diets enriched in soluble fiber improve indices of inflammation and oxidative stress in rats with Zucker fat syndrome. Pharmacol. Res. 2011, 64, 31–35. [Google Scholar] [CrossRef]
- Ma, Y.; Griffith, J.A.; Chasan-Taber, L.; Olendzki, B.C.; Jackson, E.; Stanek, E.J.; Li, W.; Pagoto, S.L.; Hafner, A.R.; Ockene, I.S. Relationship between dietary fiber and serum C-reactive protein levels. Am. J. Clin. Nutr. 2006, 83, 760–766. [Google Scholar] [CrossRef] [PubMed]
- Moyano, G.; Sáyago-Ayerdi, S.G.; Largo, C.; Caz, V.; Santamaria, M.; Tabernero, M. Potential use of dietary fibre from Hibiscus sabdariffa and Agave tequilana in obesity management. J. Funct. Foods 2016, 21, 1–9. [Google Scholar] [CrossRef]
- Dahiya, D.K.; Renuka; Puniya, M.; Shandilya, U.K.; Dhewa, T.; Kumar, N.; Kumar, S.; Puniya, A.K.; Shukla, P. Modulation of gut microbiota and its association with obesity using prebiotic fiber and probiotics: A review. Front. Microbiol. 2017, 8, 563. [Google Scholar] [CrossRef]
- Flachs, P.; Rossmeisl, M.; Bryhn, M.; Kopecky, J. Effects of N-3 polyunsaturated fatty acids on adipose tissue biology and metabolism at the cellular and molecular levels. Clin. Sci. 2009, 116, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Albracht-Schulte, K.; Kalupahana, N.S.; Ramalingam, L.; Wang, S.; Rahman, S.M.; Robert-McComb, J.; Moustaid-Moussa, N. Omega-3 fatty acids in obesity and metabolic syndrome: A mechanistic update. J. Nutr. Biochem. 2018, 58, 1–16. [Google Scholar]
- Ramírez-Moreno, E.; Arias-Rico, J.; Jiménez-Sánchez, R.C.; Estrada-Luna, D.; Jiménez-Osorio, A.S.; Zafra-Rojas, Q.Y.; Ariza-Ortega, J.A.; Flores-Chávez, O.R.; Morales-Castillejos, L.; Sandoval-Gallegos, E.M. Role of Bioactive Compounds in Obesity: Metabolic Mechanism Focused on Inflammation. Foods 2022, 11, 1232. [Google Scholar] [CrossRef]
- Grosso, G.; Laudisio, D.; Frias-Toral, E.; Barrea, L.; Muscogiuri, G.; Savastano, S.; Colao, A. Anti-Inflammatory Nutrients and Obesity-Associated Metabolic-Inflammation: State of the Art and Future Direction. Nutrients 2022, 14, 1137. [Google Scholar] [CrossRef]
- Pelczyńska, M.; Moszak, M.; Wesołek, A.; Bogdański, P. The Preventive Mechanisms of Bioactive Food Compounds against Obesity-Induced Inflammation. Antioxidants 2023, 12, 1232. [Google Scholar] [CrossRef] [PubMed]
- Lan-Lan, Z.; Liang, M.; Chuan-Geng, M.; Mei-Zhen, F.; Yan, C.; Qin, J. Effects on animal models of depression of bioactive compounds from entomogenous fungi, a novel antioxidant. Chin. J. Integr. Med. 2004, 10, 221–225. [Google Scholar] [CrossRef]
- Sarris, J.; Panossian, A.; Schweitzer, I.; Stough, C.; Scholey, A. Herbal medicine for depression, anxiety and insomnia: A review of psychopharmacology and clinical evidence. Eur. Neuropsychopharmacol. 2011, 21, 841–860. [Google Scholar] [CrossRef] [PubMed]
- Liao, L.-Y.; He, Y.-F.; Li, L.; Meng, H.; Dong, Y.-M.; Yi, F.; Xiao, P.-G. A preliminary review of studies on adaptogens: Comparison of their bioactivity in TCM with that of ginseng-like herbs used worldwide. Chin. Med. 2018, 13, 57. [Google Scholar] [CrossRef]
- Lee, G.; Bae, H. Therapeutic Effects of Phytochemicals and Medicinal Herbs on Depression. BioMed Res. Int. 2017, 2017, 6596241. [Google Scholar] [CrossRef]
- Nurzyńska-Wierdak, R. Plants with Potential Importance in Supporting the Treatment of Depression: Current Trends, and Research. Pharmaceuticals 2024, 17, 1489. [Google Scholar] [CrossRef]
- Kalueff, A.V.; Nutt, D.J. Role of GABA in anxiety and depression. Depress. Anxiety 2007, 24, 495–517. [Google Scholar] [CrossRef] [PubMed]
- Biesalski, H.-K.; Dragsted, L.O.; Elmadfa, I.; Grossklaus, R.; Müller, M.; Schrenk, D.; Walter, P.; Weber, P. Bioactive compounds: Definition and assessment of activity. Nutrition 2009, 25, 1202–1205. [Google Scholar] [CrossRef]
- Kussmann, M.; Abe Cunha, D.H.; Berciano, S. Bioactive compounds for human and planetary health. Front. Nutr. 2023, 10, 1193848. [Google Scholar] [CrossRef]
- Hoang, T.; Kim, J. Phytonutrient supplements and metabolic biomarkers of cardiovascular disease: An umbrella review of meta-analyses of clinical trials. Phytother. Res. 2021, 35, 4171–4182. [Google Scholar] [CrossRef]
- Murawska-Ciałowicz, E. Adipose tissue—Morphological and biochemical characteristic of different depots. Postepy Hig. Med. Dosw. 2017, 71, 466–484. [Google Scholar] [CrossRef] [PubMed]
- Kolb, H. Obese visceral fat tissue inflammation: From protective to detrimental? BMC Med. 2022, 20, 494. [Google Scholar] [CrossRef]
- Balistreri, C.R.; Caruso, C.; Candore, G. The role of adipose tissue and adipokines in obesity-related inflammatory diseases. Mediat. Inflamm. 2010, 2010, 802078. [Google Scholar] [CrossRef]
- Skowrońska, B.; Fichna, M.; Fichna, B. The role of adipose tissue in the endocrine system. Endokrynol. Otył. Zab. Przem. Mat. 2005, 1, 21–29. [Google Scholar]
- Pérez-Torres, I.; Castrejón-Téllez, V.; Soto, M.E.; Rubio-Ruiz, M.E.; Manzano-Pech, L.; Guarner-Lans, V. Oxidative Stress, Plant Natural Antioxidants, and Obesity. Int. J. Mol. Sci. 2021, 22, 1786. [Google Scholar] [CrossRef]
- Kuntz, S.; Kunz, C.; Domann, E.; Würdemann, N.; Unger, F.; Römpp, A.; Rudloff, S. Inhibition of Low-Grade Inflammation by Anthocyanins after Microbial Fermentation In vitro. Nutrients 2016, 8, 411. [Google Scholar] [CrossRef] [PubMed]
- Williamson, G. The Role of Polyphenols in Modern Nutrition. Nutr. Bull. 2017, 42, 226–235. [Google Scholar] [CrossRef]
- Boccellino, M.; Donniacuo, M.; Bruno, F.; Rinaldi, B.; Quagliuolo, L.; Ambruosi, M.; Pace, S.; De Rosa, M.; Olgaç, A.; Banoglu, E.; et al. Protective Effect of Piceatannol and Bioactive Stilbene Derivatives against Hypoxia-Induced Toxicity in H9c2 Cardiomyocytes and Structural Elucidation as 5-LOX Inhibitors. Eur. J. Med. Chem. 2019, 180, 637–647. [Google Scholar] [CrossRef]
- Bravo, L. Polyphenols: Chemistry, Dietary Sources, Metabolism, and Nutritional Significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef] [PubMed]
- Xu, D.-P.; Li, Y.; Meng, X.; Zhou, T.; Zhou, Y.; Zheng, J.; Zhang, J.-J.; Li, H.-B. Natural Antioxidants in Foods and Medicinal Plants: Extraction, Assessment and Resources. Int. J. Mol. Sci. 2017, 18, 96. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Moustaid-Moussa, N.; Chen, L.; Mo, H.; Shastri, A.; Su, R.; Bapat, P.; Kwun, I.; Shen, C.-L. Novel insights of dietary polyphenols and obesity. J. Nutr. Biochem. 2014, 25, 1–18. [Google Scholar] [CrossRef]
- Kowalska, K.; Olejnik, A.; Rychlik, J.; Grajek, W. Cranberries (Oxycoccus quadripetalus) inhibit adipogenesis and lipogenesis in 3T3-L1 cells. Food Chem. 2014, 148, 246–252. [Google Scholar] [CrossRef] [PubMed]
- Boqué, N.; de la Iglesia, R.; de la Garza, A.L.; Milagro, F.I.; Olivares, M.; Bañuelos, O.; Soria, A.C.; Rodríguez-Sánchez, S.; Martínez, J.A.; Campión, J. Prevention of diet-induced obesity by apple polyphenols in Wistar rats through regulation of adipocyte gene expression and DNA methylation patterns. Mol. Nutr. Food Res. 2013, 57, 1473–1478. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; He, Z.; Ma, N.; Chen, Z.-Y. Beneficial Effects of Dietary Polyphenols on High-Fat Diet-Induced Obesity Linking with Modulation of Gut Microbiota. J. Agric. Food Chem. 2020, 68, 33–47. [Google Scholar] [CrossRef]
- Singh, M.; Thrimawithana, T.; Shukla, R.; Adhikari, B. Managing obesity through natural polyphenols: A review. Future Foods 2020, 1–2, 100002. [Google Scholar] [CrossRef]
- Zhao, Y.; Chen, B.; Shen, J.; Wan, L.; Zhu, Y.; Yi, T.; Xiao, Z. The Beneficial Effects of Quercetin, Curcumin, and Resveratrol in Obesity. Oxidative Med. Cell. Longev. 2017, 2017, 1459497. [Google Scholar] [CrossRef]
- Jin, T.; Song, Z.; Weng, J.; Fantus, I.G. Curcumin and other dietary polyphenols: Potential mechanisms of metabolic actions and therapy for diabetes and obesity. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E201–E205. [Google Scholar] [CrossRef] [PubMed]
- Nabavi, S.F.; Russo, G.L.; Daglia, M.; Nabavi, S.M. Role of quercetin as an alternative for obesity treatment: You are what you eat! Food Chem. 2015, 179, 305–310. [Google Scholar] [CrossRef]
- Ahn, J.; Lee, H.; Kim, S.; Park, J.; Ha, T. The anti-obesity effect of quercetin is mediated by the AMPK and APK signaling pathways. Biochem. Biophys. Res. Commun. 2008, 373, 545–549. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Li, X.F.; Gao, L.; Zhang, Y.O.; Cai, G.P. Suppressive effects of quercetin-3-O-(6”-Feruloyl)-β-D-galactopyranoside on adipogenesis in 3T3–L1 preadipocytes through down-regulation of PPARγ and C/EBPα expression. Phytother. Res. 2012, 26, 438–444. [Google Scholar] [CrossRef]
- Overman, A.; Chuang, C.-C.; McIntosh, M. Quercetin attenuates inflammation in human macrophages and adipocytes exposed to macrophage-conditioned media. Int. J. Obes. 2011, 35, 1165–1172. [Google Scholar] [CrossRef] [PubMed]
- Comalada, M.; Ballester, I.; Bailón, E.; Sierra, S.; Xaus, J.; Gálvez, J.; de Medina, F.S.; Zarzuelo, A. Inhibition of pro-inflammatory markers in primary bone marrow-derived mouse macrophages by naturally occurring flavonoids: Analysis of the structure-activity relationship. Biochem. Pharmacol. 2006, 72, 1010–1021. [Google Scholar] [CrossRef] [PubMed]
- Chuang, C.-C.; Shen, W.; Chen, H.; Xie, G.; Jia, W.; Chung, S.; McIntosh, M.K. Differential effects of grape powder and its extract on glucose tolerance and chronic inflammation in high-fat-fed obese mice. J. Agric. Food Chem. 2012, 60, 12458–12468. [Google Scholar] [CrossRef]
- Chuang, C.-C.; Martinez, K.; Xie, G.; Kennedy, A.; Bumrungpert, A.; Overman, A.; Jia, W.; McIntosh, M.K. Quercetin is equally or more effective than resveratrol in attenuating tumor necrosis factor-α-mediated inflammation and insulin resistance in primary human adipocytes. Am. J. Clin. Nutr. 2010, 92, 1511–1521. [Google Scholar] [CrossRef]
- Pfeuffer, M.; Auinger, A.; Bley, U.; Kraus-Stojanowic, I.; Laue, C.; Winkler, P.; Rüfer, C.E.; Frank, J.; Bösch-Saadatmandi, C.; Rimbach, G.; et al. Effect of quercetin on traits of the metabolic syndrome, endothelial function and inflammation in men with different APOE isoforms. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 403–409. [Google Scholar] [CrossRef]
- Funakoshi, T.; Kanzaki, N.; Otsuka, Y.; Izumo, T.; Shibata, H.; Machida, S. Quercetin inhibits adipogenesis of muscle progenitor cells in vitro. Biochem. Biophys. Rep. 2018, 13, 39–44. [Google Scholar] [CrossRef]
- Kawabata, K.; Kawai, Y.; Terao, J. Suppressive effect of quercetin on acute stress-induced hypothalamic-pituitary-adrenal axis response in Wistar rats. J. Nutr. Biochem. 2010, 21, 374–380. [Google Scholar] [CrossRef]
- Merzoug, S.; Toumi, M.L.; Tahraoui, A. Quercetin mitigates Adriamycin-induced anxiety- and depression-like behaviors, immune dysfunction, and brain oxidative stress in rats. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2014, 387, 921–933. [Google Scholar] [CrossRef]
- Lee, B.; Yeom, M.; Shim, I.; Lee, H.; Hahm, D.-H. Protective Effects of Quercetin on Anxiety-Like Symptoms and Neuroinflammation Induced by Lipopolysaccharide in Rats. Evid. Based Complement. Alternat. Med. 2020, 2020, 4892415. [Google Scholar] [CrossRef]
- Kageyama, A.; Sakakibara, H.; Zhou, W.; Yoshioka, M.; Ohsumi, M.; Shimoi, K.; Yokogoshi, H. Genistein Regulated Serotonergic Activity in the Hippocampus of Ovariectomized Rats under Forced Swimming Stress. Biosci. Biotechnol. Biochem. 2010, 74, 2005–2010. [Google Scholar] [CrossRef]
- Rockwood, S.; Mason, D.; Lord, R.; Lamar, P.; Prozialeck, W.; Al-Nakkash, L. Genistein diet improves body weight, serum glucose and triglyceride levels in both male and female ob/ob mice. Diabetes Metab. Syndr. Obes. 2019, 12, 2011–2021. [Google Scholar] [CrossRef]
- Kim, H.-K.; Nelson-Dooley, C.; Della-Fera, M.A.; Yang, J.-Y.; Zhang, W.; Duan, J.; Hartzell, D.L.; Hamrick, M.W.; Baile, C.A. Genistein Decreases Food Intake, Body Weight, and Fat Pad Weight and Causes Adipose Tissue Apoptosis in Ovariectomized Female Mice. J. Nutr. 2006, 136, 409–414. [Google Scholar] [CrossRef]
- Chen, X.; Xie, J.; Tan, Q.; Li, H.; Lu, J.; Zhang, X. Genistein improves systemic metabolism and enhances cold resistance by promoting adipose tissue beiging. Biochem. Biophys. Res. Commun. 2021, 558, 154–160. [Google Scholar] [CrossRef]
- Banyś, K.; Stawarska, A.; Wyrębiak, R.; Bielecki, W.; Bobrowska-Korczak, B. The Effect of Genistein Supplementation on Cholesterol Oxidation Products and Fatty Acid Profiles in Serums of Rats with Breast Cancer. Foods 2022, 11, 605. [Google Scholar] [CrossRef] [PubMed]
- Atteritano, M.; Mazzaferro, S.; Bitto, A.; Cannata, M.L.; D’Anna, R.; Squadrito, F.; Macrì, I.; Frisina, A.; Frisina, N.; Bagnato, G. Genistein effects on quality of life and depression symptoms in osteopenic postmenopausal women: A 2-year randomized, double-blind, controlled study. Osteoporos. Int. 2014, 25, 1123–1129. [Google Scholar] [CrossRef]
- Gupta, G.; Jia Jia, T.; Yee Woon, L.; Kumar Chellappan, D.; Candasamy, M.; Dua, K. Pharmacological Evaluation of Antidepressant-Like Effect of Genistein and Its Combination with Amitriptyline: An Acute and Chronic Study. Adv. Pharmacol. Sci. 2015, 2015, 164943. [Google Scholar] [CrossRef]
- Zarmouh, N.O.; Messeha, S.S.; Elshami, F.M.; Soliman, K.F.A. Evaluation of the Isoflavone Genistein as Reversible Human Monoamine Oxidase-A and -B Inhibitor. Evid.-Based Complement. Altern. Med. 2016, 2016, 1423052. [Google Scholar] [CrossRef]
- Diepvens, K.; Westerterp, K.R.; Westerterp-Plantenga, M.S. Obesity and thermogenesis related to the consumption of caffeine, ephedrine, capsaicin, and green tea. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2007, 292, R77–R85. [Google Scholar] [CrossRef]
- Xia, N.; Daiber, A.; Förstermann, U.; Li, H. Antioxidant effects of resveratrol in the cardiovascular system. Br. J. Pharmacol. 2017, 174, 1633–1646. [Google Scholar] [CrossRef] [PubMed]
- Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; García-Almagro, F.J.; Avilés-Plaza, F.; Parra, S.; Yáñez-Gascón, M.J.; Ruiz-Ros, J.A.; García-Conesa, M.T.; Tomás-Barberán, F.A.; et al. Consumption of a grape extract supplement containing resveratrol decreases oxidized LDL and ApoB in patients undergoing primary prevention of cardiovascular disease: A triple-blind, 6-month follow-up, placebo-controlled, randomized trial. Mol. Nutr. Food Res. 2012, 56, 810–821. [Google Scholar] [CrossRef]
- Yang, J.-Y.; Della-Fera, M.A.; Rayalam, S.; Ambati, S.; Hartzell, D.L.; Park, H.J.; Baile, C.A. Enhanced inhibition of adipogenesis and induction of apoptosis in 3T3-L1 adipocytes with combinations of resveratrol and quercetin. Life Sci. 2008, 82, 1032–1039. [Google Scholar] [CrossRef]
- Chen, S.; Xiao, X.; Feng, X.; Li, W.; Zhou, N.; Zheng, L.; Sun, Y.; Zhang, Z.; Zhu, W. Resveratrol induces Sirt1-dependent apoptosis in 3T3-L1 preadipocytes by activating AMPK and suppressing AKT activity and survivin expression. J. Nutr. Biochem. 2012, 23, 1100–1112. [Google Scholar] [CrossRef] [PubMed]
- Pang, W.-J.; Xiong, Y.; Zhang, Z.; Wei, N.; Chen, N.; Yang, G.-S. Lentivirus-mediated Sirt1 shRNA and resveratrol independently induce porcine preadipocyte apoptosis by canonical apoptotic pathway. Mol. Biol. Rep. 2013, 40, 129–139. [Google Scholar] [CrossRef]
- Alberdi, G.; Rodríguez, V.M.; Miranda, J.; Macarulla, M.T.; Arias, N.; Andrés-Lacueva, C.; Portillo, M.P. Changes in white adipose tissue metabolism induced by resveratrol in rats. Nutr. Metab. 2011, 8, 29. [Google Scholar] [CrossRef]
- Nagao, K.; Jinnouchi, T.; Kai, S.; Yanagita, T. Effect of dietary resveratrol on the metabolic profile of nutrients in obese OLETF rats. Lipids Health Dis. 2013, 12, 8. [Google Scholar] [CrossRef] [PubMed]
- Albani, D.; Polito, L.; Signorini, A.; Forloni, G. Neuroprotective properties of resveratrol in different neurodegenerative disorders. Biofactors 2010, 36, 370–376. [Google Scholar] [CrossRef]
- Gu, Z.; Chu, L.; Han, Y. Therapeutic effect of resveratrol on mice with depression. Exp. Ther. Med. 2019, 18, 3015–3020. [Google Scholar] [CrossRef]
- Ali, S.H.; Madhana, R.M.; Athira, K.V.; Kasala, E.R.; Bodduluru, L.N.; Pitta, S.; Mahareddy, J.R.; Lahkar, M. Resveratrol ameliorates depressive-like behavior in repeated corticosterone-induced depression in mice. Steroids 2015, 101, 37–42. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Zhang, Q.; Cai, Y.; Sun, D.; He, X.; Wang, L.; Yu, D.; Li, X.; Xiong, X.; Xu, H.; et al. Resveratrol counteracts lipopolysaccharide-induced depressive-like behaviors via enhanced hippocampal neurogenesis. Oncotarget 2016, 7, 56045–56059. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.S.; Zhang, J.; Zhang, L.; Huang, J.; Wang, Y. Mechanisms of body weight reduction and metabolic syndrome alleviation by tea. Mol. Nutr. Food Res. 2016, 60, 160–174. [Google Scholar] [CrossRef]
- Gu, Q.; Xia, L.; Du, Q.; Shao, Y.; He, J.; Wu, P.; Liang, L.; Shen, X. The therapeutic role and potential mechanism of EGCG in obesity-related precocious puberty as determined by integrated metabolomics and network pharmacology. Front. Endocrinol. 2023, 14, 1159657. [Google Scholar] [CrossRef] [PubMed]
- Peairs, A.; Dai, R.; Gan, L.; Shimp, S.; Rylander, M.N.; Li, L.; Reilly, C.M. Epigallocatechin-3-gallate (EGCG) attenuates inflammation in MRL/lpr mouse mesangial cells. Cell. Moth. Immunol. 2010, 7, 123–132. [Google Scholar] [CrossRef] [PubMed]
- Lee, B.; Shim, I.; Lee, H.; Hahm, D.-H. Effects of Epigallocatechin Gallate on Behavioral and Cognitive Impairments, Hypothalamic–Pituitary–Adrenal Axis Dysfunction, and Alterations in Hippocampal BDNF Expression Under Single Prolonged Stress. J. Med. Food 2018, 21, 979–989. [Google Scholar] [CrossRef]
- El-Missiry, M.A.; Othman, A.I.; El-Sawy, M.R.; Lebede, M.F. Neuroprotective effect of epigallocatechin-3-gallate (EGCG) on radiation-induced damage and apoptosis in the rat hippocampus. Int. J. Radiat. Biol. 2018, 94, 798–808. [Google Scholar] [CrossRef]
- Li, F.; Gao, C.; Yan, P.; Zhang, M.; Wang, Y.; Hu, Y.; Wu, X.; Wang, X.; Sheng, J. EGCG Reduces Obesity and White Adipose Tissue Gain Partly Through AMPK Activation in Mice. Front. Pharmacol. 2018, 9, 1366. [Google Scholar] [CrossRef]
- Hwang, Y.P.; Choi, J.H.; Han, E.H.; Kim, H.G.; Wee, J.-H.; Jung, K.O.; Jung, K.H.; Kwon, K.; Jeong, T.C.; Chung, Y.C.; et al. Purple sweet potato anthocyanins attenuate hepatic lipid accumulation through activating adenosine monophosphate–activated protein kinase in human HepG2 cells and obese mice. Nutr. Res. 2011, 31, 896–906. [Google Scholar] [CrossRef] [PubMed]
- Sato, D.; Kusunoki, M.; Seino, N.; Nishina, A.; Feng, Z.; Tsutsumi, K.; Nakamura, T. Black soybean extract reduces fatty acid contents in subcutaneous, but not in visceral adipose triglyceride in high-fat fed rats. Int. J. Food Sci. Nutr. 2015, 66, 539–545. [Google Scholar] [CrossRef]
- Zhang, H.; Koes, R.; Shang, H.; Fu, Z.; Wang, L.; Dong, X.; Zhang, J.; Passeri, V.; Li, Y.; Jiang, H.; et al. Identification and functional analysis of three new anthocyanin R2R3-MYB genes in Petunia. Plant Direct 2019, 3, e00114. [Google Scholar] [CrossRef]
- Habtemariam, S.; Varghese, G.K. The antidiabetic therapeutic potential of dietary polyphenols. Curr. Pharm. Biotechnol. 2014, 15, 391–400. [Google Scholar] [CrossRef] [PubMed]
- Tucakovic, L.; Colson, N.; Santhakumar, A.B.; Kundur, A.R.; Shuttleworth, M.; Singh, I. The effects of anthocyanins on body weight and expression of adipocyte’s hormones: Leptin and adiponectin. J. Funct. Foods 2018, 45, 173–180. [Google Scholar] [CrossRef]
- Yildiz, E.; Guldas, M.; Ellergezen, P.; Acar, A.G.; Gurbuz, O. Obesity-associated Pathways of Anthocyanins. Food Sci. Technol. 2021, 41, 1–13. [Google Scholar] [CrossRef]
- Chen, Z.; Wang, C.; Pan, Y.; Gao, X.; Chen, H. Hypoglycemic and hypolipidemic effects of anthocyanins extract from black soybean seed coat in high fat diet and streptozotocin-induced diabetic mice. Food Funct. 2018, 9, 426–439. [Google Scholar] [CrossRef]
- Garcia, D.; Shaw, R.J. AMPK: Mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance. Mol. Cell 2017, 66, 789–800. [Google Scholar] [CrossRef]
- Hidalgo, M.; Oruna-Concha, M.J.; Kolida, S.; Walton, G.E.; Kallithraka, S.; Spencer, J.P.E.; de Pascual-Teresa, S. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J. Agric. Food Chem. 2012, 60, 3882–3890. [Google Scholar] [CrossRef]
- Jamar, G.; Estadella, D.; Pisani, L.P. Contribution of anthocyanin-rich foods in obesity control through gut microbiota interactions. Biofactors 2017, 43, 507–516. [Google Scholar] [CrossRef]
- Wybraniec, S.; Mizrahi, Y. Fruit flesh betacyanin pigments in Hylocereus cacti. J. Agric. Food Chem. 2002, 50, 6086–6089. [Google Scholar] [CrossRef] [PubMed]
- Wybraniec, S.; Platzner, I.; Geresh, S.; Gottlieb, H.E.; Haimberg, M.; Mogilnitzki, M.; Mizrahi, Y. Betacyanins from vine cactus Hylocereus polyrhizus. Phytochemistry 2001, 58, 1209–1212. [Google Scholar] [CrossRef]
- Nakamura, K.; Fuster, J.J.; Walsh, K. Adipokines: A link between obesity and cardiovascular disease. J. Cardiol. 2014, 63, 250–259. [Google Scholar] [CrossRef]
- Ahmadi, H.; Nayeri, Z.; Minuchehr, Z.; Sabouni, F.; Mohammadi, M. Betanin purification from red beetroots and evaluation of its antioxidant and anti-inflammatory activity on LPS-activated microglial cells. PLoS ONE 2020, 15, e0233088. [Google Scholar] [CrossRef] [PubMed]
- Khalili, R.M.A.; Norhayati, A.H.; Rokiah, M.Y.; Asmah, R.; Muskinah, M.S.; Manaf, A.A. Hypocholesterolemic effect of red pitaya (Hylocereus sp.) on hypercholesterolemia induced rats. Int. Food Res. J. 2009, 16, 431–440. [Google Scholar]
- Lugo-Radillo, A.; Delgado-Enciso, I.; Rodriguez-Hernandez, A.; Peña-Beltran, E.; Martinez-Martinez, R.; Galvan-Salazar, H. Inhibitory effect of betanin from Hylocereus ocamponis against steatohepatitis in mice fed a high-fat diet. Nat. Prod. Commun. 2020, 15, 1934578X20932013. [Google Scholar] [CrossRef]
- Sakihama, Y.; Maeda, M.; Hashimoto, M.; Tahara, S.; Hashidoko, Y. Beetroot betalain inhibits peroxynitrite-mediated tyrosine nitration and DNA strand cleavage. Free Radic. Res. 2012, 46, 93–99. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Maqueo, A.; Soccio, M.; Cano, M.P. In vitro antioxidant capacity of Opuntia spp. fruits measured by the LOX-FL method and its high sensitivity towards betalains. Plant Foods Hum. Nutr. 2021, 76, 354–362. [Google Scholar] [CrossRef]
- Smeriglio, A.; Bonasera, S.; Germanò, M.P.; D’Angelo, V.; Barreca, D.; Denaro, M.; Monforte, M.T.; Galati, E.M.; Trombetta, D. Opuntia ficus-indica (L.) Mill. fruit as source of betalains with antioxidant, cytoprotective, and anti-angiogenic properties. Phytother. Res. 2019, 33, 1526–1537. [Google Scholar] [CrossRef]
- Khoo, H.E.; He, X.; Tang, Y.; Li, Z.; Li, C.; Zeng, Y.; Tang, J.; Sun, J. Betacyanins and anthocyanins in pulp and peel of red pitaya (Hylocereus polyrhizus cv. Jindu), inhibition of oxidative stress, lipid reducing, and cytotoxic effects. Front. Nutr. 2022, 9, 894438. [Google Scholar] [CrossRef] [PubMed]
- Escribano, J.; Cabanes, J.; Jiménez-Atiénzar, M.; Ibañez-Tremolada, M.; Gómez-Pando, L.R.; García-Carmona, F.; Gandía-Herrero, F. Characterization of betalains, saponins and antioxidant power in differently colored quinoa (Chenopodium quinoa) varieties. Food Chem. 2017, 234, 285–294. [Google Scholar] [CrossRef]
- Martínez-Rodríguez, P.; Guerrero-Rubio, M.A.; Henarejos-Escudero, P.; García-Carmona, F.; Gandía-Herrero, F. Health-promoting potential of betalains in vivo and their relevance as functional ingredients: A review. Trends Food Sci. Technol. 2022, 122, 66–82. [Google Scholar] [CrossRef]
- Szafrański, T. Herbal remedies in depression—State of the art. Psychiatr. Pol. 2014, 48, 59–73. [Google Scholar] [CrossRef]
- Pohl, F.; Kong Thoo Lin, P. The potential use of plant natural products and plant extracts with antioxidant properties for the prevention/treatment of neurodegenerative diseases: In vitro, in vivo and clinical trials. Molecules 2018, 23, 3283. [Google Scholar] [CrossRef] [PubMed]
- Fathinezhad, Z.; Sewell, R.D.E.; Lorigooini, Z.; Rafieian-Kopaei, M. Depression and treatment with effective herbs. Curr. Pharm. Des. 2019, 25, 738–745. [Google Scholar] [CrossRef]
- Bruni, O.; Ferini-Strambi, L.; Giacomoni, E.; Pellegrino, P. Herbal remedies and their possible effect on the GABAergic system and sleep. Nutrients 2021, 13, 530. [Google Scholar] [CrossRef] [PubMed]
- Rai, D.; Bhatia, G.; Palit, G.; Pal, R.; Singh, S.; Singh, H.K. Adaptogenic effect of Bacopa monniera (Brahmi). Pharmacol. Biochem. Behav. 2003, 75, 823–830. [Google Scholar] [CrossRef]
- Novío, S.; Núñez, M.J.; Amigo, G.; Freire-Garabal, M. Effects of fluoxetine on the oxidative status of peripheral blood leucocytes of restraint-stressed mice. Basic Clin. Pharmacol. Toxicol. 2011, 109, 365–371. [Google Scholar] [CrossRef] [PubMed]
- Rabiei, Z.; Rabiei, S. A review on antidepressant effect of medicinal plants. Bangladesh J. Pharmacol. 2017, 12, 1–11. [Google Scholar] [CrossRef]
- Soyman, Z.; Kelekçi, S.; Sal, V.; Şevket, O.; Bayındır, N.; Uzun, H. Effects of apigenin on experimental ischemia/reperfusion injury in the rat ovary. Balkan Med. J. 2017, 34, 444–449. [Google Scholar] [CrossRef]
- Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E.; et al. The therapeutic potential of apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef]
- Weng, L.; Guo, X.; Li, Y.; Yang, X.; Han, Y. Apigenin reverses depression-like behavior induced by chronic corticosterone treatment in mice. Eur. J. Pharmacol. 2016, 774, 50–54. [Google Scholar] [CrossRef]
- Li, R.; Zhao, D.; Qu, R.; Fu, Q.; Ma, S. The effects of apigenin on lipopolysaccharide-induced depressive-like behavior in mice. Neurosci. Lett. 2015, 594, 17–22. [Google Scholar] [CrossRef]
- Zhang, X.; Bu, H.; Jiang, Y.; Sun, G.; Jiang, R.; Huang, X.; Duan, H.; Huang, Z.; Wu, Q. The antidepressant effects of apigenin are associated with the promotion of autophagy via the mTOR/AMPK/ULK1 pathway. Mol. Med. Rep. 2019, 20, 2867–2874. [Google Scholar] [CrossRef] [PubMed]
- Yi, L.-T.; Li, J.; Li, H.-C.; Su, D.-X.; Quan, X.-B.; He, X.-C.; Wang, X.-H. Antidepressant-like behavioral, neurochemical and neuroendocrine effects of naringenin in the mouse repeated tail suspension test. Prog. Neuropsychopharmacol. Biol. Psychiatry 2012, 39, 175–181. [Google Scholar] [CrossRef] [PubMed]
- Yi, L.-T.; Liu, B.-B.; Li, J.; Luo, L.; Liu, Q.; Geng, D.; Tang, Y.; Xia, Y.; Wu, D. BDNF signaling is necessary for the antidepressant-like effect of naringenin. Prog. Neuropsychopharmacol. Biol. Psychiatry 2014, 48, 135–141. [Google Scholar] [CrossRef]
- Bansal, Y.; Singh, R.; Saroj, P.; Sodhi, R.K.; Kuhad, A. Naringenin protects against oxido-inflammatory aberrations and altered tryptophan metabolism in olfactory bulbectomized mice model of depression. Toxicol. Appl. Pharmacol. 2018, 355, 257–268. [Google Scholar] [CrossRef] [PubMed]
- Barati, N.; Momtazi-Borojeni, A.A.; Majeed, M.; Sahebkar, A. Potential therapeutic effects of curcumin in gastric cancer. J. Cell. Physiol. 2019, 234, 2317–2328. [Google Scholar] [CrossRef] [PubMed]
- Okereke, O.I.; Cook, N.R.; Albert, C.M.; Van Denburgh, M.; Buring, J.E.; Manson, J.E. Effect of long-term supplementation with folic acid and B vitamins on risk of depression in older women. Br. J. Psychiatry 2015, 206, 324–331. [Google Scholar] [CrossRef]
- Kulkarni, S.K.; Bhutani, M.K.; Bishnoi, M. Antidepressant activity of curcumin: Involvement of serotonin and dopamine system. Psychopharmacology 2008, 201, 435–442. [Google Scholar] [CrossRef]
- Ng, Q.X.; Koh, S.S.H.; Chan, H.W.; Ho, C.Y.X. Clinical use of curcumin in depression: A meta-analysis. J. Am. Med. Dir. Assoc. 2017, 18, 503–508. [Google Scholar] [CrossRef]
- Bhutani, M.K.; Bishnoi, M.; Kulkarni, S.K. Anti-depressant like effect of curcumin and its combination with piperine in unpredictable chronic stress-induced behavioral, biochemical and neurochemical changes. Pharmacol. Biochem. Behav. 2009, 92, 39–43. [Google Scholar] [CrossRef]
- Nazemi, H.; Mirzaei, M.; Jafari, E. Antidepressant activity of curcumin by monoamine oxidase–A inhibition. Adv. J. Chem.-Sect. B 2019, 1, 3–9. [Google Scholar] [CrossRef]
- Andrade, C. A Critical Examination of Studies on Curcumin for Depression. J. Clin. Psychiatry 2014, 75, e1110–e1112. [Google Scholar] [CrossRef] [PubMed]
- Fusar-Poli, L.; Vozza, L.; Gabbiadini, A.; Vanella, A.; Concas, I.; Tinacci, S.; Petralia, A.; Signorelli, M.S.; Aguglia, E. Curcumin for depression: A meta-analysis. Crit. Rev. Food Sci. Nutr. 2020, 60, 2643–2653. [Google Scholar] [CrossRef] [PubMed]
- Mao, Q.-Q.; Xian, Y.-F.; Ip, S.-P.; Che, C.-T. Involvement of serotonergic system in the antidepressant-like effect of piperine. Prog. Neuropsychopharmacol. Biol. Psychiatry 2011, 35, 1144–1147. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.A.; Hong, S.S.; Han, X.H.; Hwang, J.S.; Oh, G.J.; Lee, K.S.; Lee, M.K.; Hwang, B.Y.; Ro, J.S. Piperine from the Fruits of Piper longum with Inhibitory Effect on Monoamine Oxidase and Antidepressant-Like Activity. Chem. Pharm. Bull. 2005, 53, 832–835. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Wang, C.; Li, W.; Koike, K.; Nikaido, T.; Wang, M.-W. Antidepressant-like effects of piperine and its derivative, antiepilepsirine. J. Asian Nat. Prod. Res. 2007, 9, 421–430. [Google Scholar] [CrossRef]
- Mao, Q.-Q.; Huang, Z.; Ip, S.-P.; Xian, Y.-F.; Che, C.-T. Role of 5-HT1A and 5-HT1B receptors in the antidepressant-like effect of piperine in the forced swim test. Neurosci. Lett. 2011, 504, 181–184. [Google Scholar] [CrossRef]
- Mao, Q.-Q.; Huang, Z.; Zhong, X.-M.; Xian, Y.-F.; Ip, S.-P. Piperine reverses the effects of corticosterone on behavior and hippocampal BDNF expression in mice. Neurochem. Int. 2014, 74, 36–41. [Google Scholar] [CrossRef]
- Li, S.; Wang, C.; Wang, M.; Li, W.; Matsumoto, K.; Tang, Y. Antidepressant-like effects of piperine in chronic mild stress treated mice and its possible mechanisms. Life Sci. 2007, 80, 1373–1381. [Google Scholar] [CrossRef] [PubMed]
- Ishisaka, M.; Kakefuda, K.; Yamauchi, M.; Tsuruma, K.; Shimazawa, M.; Tsuruta, A.; Hara, H. Luteolin shows an antidepressant-like effect via suppressing endoplasmic reticulum stress. Biol. Pharm. Bull. 2011, 34, 1481–1486. [Google Scholar] [CrossRef]
- Gupta, G.; Tiwari, J.; Dahiya, R.; Sharma, R.K.; Mishra, A.; Dua, K. Recent updates on neuropharmacological effects of luteolin. EXCLI J. 2018, 17, Doc211. [Google Scholar] [CrossRef]
- Lee, J.K.; Kim, S.Y.; Kim, Y.S.; Lee, W.-H.; Hwang, D.H.; Lee, J.Y. Suppression of the TRIF-dependent signaling pathway of Toll-like receptors by luteolin. Biochem. Pharmacol. 2009, 77, 1391–1400. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.-W.; Wu, M.-J.; Liu, I.Y.-C.; Su, J.-D.; Yen, J.-H. Neurotrophic and cytoprotective action of luteolin in PC12 cells through ERK-dependent induction of Nrf2-driven HO-1 expression. J. Agric. Food Chem. 2010, 58, 4477–4486. [Google Scholar] [CrossRef]
- Sur, B.; Lee, B. Luteolin reduces fear, anxiety, and depression in rats with post-traumatic stress disorder. Anim. Cells Syst. 2022, 26, 174–182. [Google Scholar] [CrossRef]
- Zhu, S.; Lei, S.; Zhou, S.; Jin, L.; Zeng, S.; Jiang, H.; Zhou, H. Luteolin shows antidepressant-like effect by inhibiting and downregulating plasma membrane monoamine transporter (PMAT, Slc29a4). J. Funct. Foods 2019, 54, 440–448. [Google Scholar] [CrossRef]
- Shi, X.; Fu, Y.; Zhang, S.; Ding, H.; Chen, J. Baicalin attenuates subarachnoid hemorrhagic brain injury by modulating blood–brain barrier disruption, inflammation, and oxidative damage in mice. Oxidative Med. Cell. Longev. 2017, 2017, 1401790. [Google Scholar] [CrossRef] [PubMed]
- Zuo, D.; Lin, L.; Liu, Y.; Wang, C.; Xu, J.; Sun, F.; Li, L.; Li, Z.; Wu, Y. Baicalin attenuates ketamine-induced neurotoxicity in the developing rats: Involvement of PI3K/Akt and CREB/BDNF/Bcl-2 pathways. Neurotox. Res. 2016, 30, 159–172. [Google Scholar] [CrossRef]
- Miller, A.H.; Raison, C.L. The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2016, 16, 22–34. [Google Scholar] [CrossRef]
- Lu, Y.; Sun, G.; Yang, F.; Guan, Z.; Zhang, Z.; Zhao, J.; Liu, Y.; Chu, L.; Pei, L. Baicalin regulates depression behavior in mice exposed to chronic mild stress via the Rac/LIMK/cofilin pathway. Biomed. Pharmacother. 2019, 116, 109054. [Google Scholar] [CrossRef]
- Yu, H.-Y.; Yin, Z.-J.; Yang, S.-J.; Ma, S.-P. Baicalin reverses AMPA receptor expression and neuron apoptosis in chronic unpredictable mild stress rats. Biochem. Biophys. Res. Commun. 2014, 451, 467–472. [Google Scholar] [CrossRef]
- Wang, D.; Ren, Y.-M.; Guo, Y.-X.; Zhang, Z.-Q.; Sui, H.; Zhang, H.-Y. The effects of baicalin in depression: Preclinical evidence construction based on meta-analysis. Front. Pharmacol. 2024, 15, 1425094. [Google Scholar] [CrossRef]
- Chen, S.; Li, Z.; Li, W.; Shan, Z.; Zhu, W. Resveratrol inhibits cell differentiation in 3T3-L1 adipocytes via activation of AMPK. Can. J. Physiol. Pharmacol. 2011, 89, 793–799. [Google Scholar]
- Zhao, L.; Chen, J.; Liu, Y.; Ding, W.; Yang, L.; Li, Y. Curcumin inhibits lipid accumulation and inflammation in 3T3-L1 cells through modulation of NF-κB signaling pathway. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158747. [Google Scholar]
- Jang, M.; Park, S.W.; Chi, S.; Cho, S.Y.; Lee, K.H.; Kim, Y.C.; Kwon, S.H. Curcumin attenuates inflammation in macrophages via NF-κB pathway inhibition. J. Nutr. Biochem. 2018, 59, 126–133. [Google Scholar]
- Wang, Y.; Liu, J.; Zhang, H.; Sun, C.; Yang, Y.; Wang, G.; Zhang, J.; Shi, Y. Epigallocatechin gallate suppresses adipocyte differentiation and lipid accumulation in 3T3-L1 cells via downregulation of PPARγ and C/EBPα. Food Funct. 2018, 9, 2090–2100. [Google Scholar]
- Kim, Y.; Lee, S.; Park, J.; Choi, S. Effects of Curcuma longa extract supplementation on obesity and depression in high-fat diet-fed female rats. J. Nutr. Biochem. 2021, 89, 108570. [Google Scholar]
- Zhang, H.; Wang, X.; Li, Y.; Chen, Z. Anthocyanin-rich acai berry extract improves metabolic and behavioral parameters in a mouse model of depression induced by chronic stress and high carbohydrate diet. Neuropharmacology 2020, 178, 108234. [Google Scholar] [CrossRef]
- Khan, M.A.; Lee, K.M.; Kim, Y.S.; Choe, S.U.; Son, D.H.; Kim, Y.N.; Kim, S.Y.; Jung, J.H. Epigallocatechin gallate ameliorates obesity and depression-like behaviors in high-fat diet-fed mice. Neuropharmacology 2019, 155, 66–77. [Google Scholar]
- Zhang, L.; Li, Y.; Liu, H.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Omega-3 fatty acids improve metabolic and neurochemical parameters in diet-induced obese rats. Prostaglandins Leukot. Essent. Fatty Acids 2020, 157, 102076. [Google Scholar]
- Singh, R.; Kumar, S.; Patel, A. Nigella sativa oil ameliorates obesity and depression-like behavior in rats: Role of anti-inflammatory and neuroprotective mechanisms. Phytother. Res. 2022, 36, 1522–1532. [Google Scholar] [CrossRef]
- Nguyen, T.T.; Tran, C.M.; Le, L.H.; Nguyen, V.H.; Le, H.M.; Nguyen, K.A. Curcumin supplementation ameliorates depression and metabolic disturbances in stressed rats. Neuroendocrinol. Lett. 2021, 42, 312–320. [Google Scholar]
- Lee, B.; Kim, C.Y. Dietary bioactive compounds for managing obesity and depression: A clinical perspective. Nutrients 2021, 13, 1254. [Google Scholar]
- Kim, S.; Lee, J.; Park, H.; Hwang, S.; Kim, H.; Kim, C.; Kim, T.; Park, S. Curcumin supplementation improves metabolic profiles and behavior in obese mice. J. Nutr. Biochem. 2018, 64, 62–71. [Google Scholar]
- Inoue, T.; Takaki, M.; Sato, H.; Fujii, M.; Matsuyama, A.; Yamashita, T.; Okamura, M.; Nakatani, K.; Nakanishi, M. Resveratrol improves insulin sensitivity and reduces adiposity in high-fat diet-fed mice. Nutr. Metab. 2019, 16, 1–10. [Google Scholar]
- Zhang, L.; Li, H.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Green tea extract modulates gut microbiota and alleviates obesity and depression in rats. Neurosci. Lett. 2020, 736, 135278. [Google Scholar]
- Li, Y.; Zhang, J.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Synergistic effects of curcumin and probiotics on obesity and mood in diet-induced obese mice. Front. Microbiol. 2021, 12, 652345. [Google Scholar]
- Wang, Q.; Liu, M.; Zhang, W.; Li, Y.; Chen, X.; Sun, H.; Ma, B.; Wang, Q.; Zhang, H.; Li, J.; et al. Combined effect of ginger extract and probiotics on metabolic and psychological parameters in obese patients: A randomized controlled trial. J. Clin. Med. 2022, 11, 2112. [Google Scholar]
- Li, H.; Zhang, J.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Phenolic compounds from berries modulate gut microbiota and SCFA production. Food Chem. 2019, 278, 95–105. [Google Scholar] [CrossRef]
- Zhao, J.; Liu, J.; Zhang, H.; Sun, C.; Yang, Y.; Wang, G.; Zhang, J.; Shi, Y. Fermentation of berry extracts enhances SCFA production and gut barrier function. J. Agric. Food Chem. 2021, 69, 1024–1033. [Google Scholar]
- Chen, L.; Wang, S.; Li, X.; Zhang, M.; Liu, Y.; Wang, J.; Ma, X. Effects of dietary polyphenols on gut microbiota and metabolic health in obese rats. Food Funct. 2020, 11, 3543–3555. [Google Scholar]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The role of short-chain fatty acids in gut-brain communication. Brain Behav. Immun. 2020, 87, 40–52. [Google Scholar]
- Kaur, S.; Singh, S.; Kumar, R.; Kumar, A.; Sharma, A.; Singh, N.; Kaur, R.; Kaur, H.; Singh, D. Curcumin ameliorates depression-like behavior and metabolic disturbances via gut microbiota modulation. Neuropharmacology 2019, 154, 137–149. [Google Scholar]
- Zhou, Y.; Zhang, J.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Curcumin supplementation alleviates depressive behaviors and modulates gut microbiota in obese rats. Neurosci. Lett. 2021, 744, 135564. [Google Scholar]
- Gao, X.; Li, H.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Synergistic effects of probiotics and herbal extracts on gut health and metabolic parameters in mice. J. Funct. Foods 2018, 45, 107–117. [Google Scholar]
- Wang, H.; Zhang, J.; Wang, X.; Liu, J.; Lin, J.; Chen, J.; Wei, H.; Wang, T. Synbiotic intervention improves metabolic health and gut microbiota in obese mice. Sci. Rep. 2020, 10, 12345. [Google Scholar]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, C.N.; Cussotto, S.; Fulling, C.; Funke, L.; et al. The microbiota–gut–brain axis. Physiol. Rev. 2020, 100, 2151–2185. [Google Scholar]
- Li, X.; Chen, X.; Sun, H.; Ma, B.; Wang, Q.; Zhang, H.; Li, J.; Zhao, Z.; Wang, X. Prebiotics and probiotics in the management of obesity and depression: A translational perspective. Front. Nutr. 2022, 9, 887654. [Google Scholar]
- Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef]
- Zupancic, S.; Lavric, Z.; Kristl, J. Stability and solubility of trans-resveratrol are strongly influenced by pH and temperature. Eur. J. Pharm. Biopharm. 2015, 93, 196–204. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; He, H.; Wang, G.; Yang, B.; Ren, W.; Ma, L.; Yu, Q. Stereospecific determination of cis- and trans-resveratrol in rat plasma by HPLC: Application to pharmacokinetic studies. Biomed. Chromatogr. 2007, 21, 257–265. [Google Scholar] [CrossRef]
- Delmas, D.; Aires, V.; Limagne, E.; Dutartre, P.; Mazue, F.; Ghiringhelli, F.; Latruffe, N. Transport, stability, and biological activity of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 48–59. [Google Scholar] [CrossRef]
- Burkon, A.; Somoza, V. Quantification of free and protein-bound trans-resveratrol metabolites and identification of trans-resveratrol-C/O-conjugated diglucuronides—Two novel resveratrol metabolites in human plasma. Mol. Nutr. Food Res. 2008, 52, 549–557. [Google Scholar] [CrossRef]
- Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov 2006, 5, 493–506. [Google Scholar] [PubMed]
- Giuliani, C.; Bucci, I.; Di Santo, S.; Rossi, C.; Grassadonia, A.; Mariotti, M.; Piantelli, M.; Monaco, F.; Napolitano, G. Resveratrol inhibits sodium/iodide symporter gene expression and function in rat thyroid cells. PLoS ONE 2014, 9, e107936. [Google Scholar]
- Giuliani, C.; Iezzi, M.; Ciolli, L.; Hysi, A.; Bucci, I.; Di Santo, S.; Rossi, C.; Zucchelli, M.; Napolitano, G. Resveratrol has anti-thyroid effects both in vitro and in vivo. Food Chem. Toxicol. 2017, 107, 237–247. [Google Scholar] [PubMed]
- Bolton, J.L.; Dunlap, T. Formation and Biological Targets of Quinones: Cytotoxic versus Cytoprotective Effects. Chem. Res. Toxicol. 2017, 30, 13–37. [Google Scholar] [CrossRef]
- Pierce, E.N.; Piyankarage, S.C.; Dunlap, T.; Litosh, V.; Siklos, M.I.; Wang, Y.T.; Thatcher, G.R. Prodrugs Bioactivated to Quinones Target NF-kappaB and Multiple Protein Networks: Identification of the Quinonome. Chem. Res. Toxicol. 2016, 29, 1151–1159. [Google Scholar] [CrossRef] [PubMed]
- Piver, B.; Fer, M.; Vitrac, X.; Merillon, J.M.; Dreano, Y.; Berthou, F.; Lucas, D. Involvement of cytochrome P450 1A2 in the biotransformation of trans-resveratrol in human liver microsomes. Biochem. Pharm. 2004, 68, 773–782. [Google Scholar] [CrossRef] [PubMed]
- Atmaca, N.; Yıldırım, E.; Güner, B.; Kabakçi, R.; Bilmen, F. Effect of Resveratrol on Hematological and Biochemical Alterations in Rats Exposed to Fluoride. BioMed Res. Int. 2014, 2014, 698628. [Google Scholar] [CrossRef]
- Wang, Y.; Cui, H.; Niu, F.; Liu, S.-L.; Li, Y.; Zhang, L.-M.; Du, H.-B.; Zhao, Z.-G.; Niu, C.-Y. Effect of Resveratrol on Blood Rheological Properties in LPS-Challenged Rats. Front. Physiol. 2018, 9, 1202. [Google Scholar] [CrossRef]
- Muñoz, O.; Bustamante, S. Pharmacological Properties of Resveratrol. A Pre-Clinical and Clinical Review. Biochem. Pharmacol. Open Access 2015, 4, 1000184. [Google Scholar] [CrossRef]
- Johnson, W.D.; Morrissey, R.L.; Usborne, A.L.; Kapetanovic, I.; Crowell, J.A.; Muzzio, M.; McCormick, D.L. Subchronic oral toxicity and cardiovascular safety pharmacology studies of resveratrol, a naturally occurring polyphenol with cancer preventive activity. Food Chem. Toxicol. 2011, 49, 3319–3327. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Zhao, M.; Zhou, Y.; Wang, C.; Yuan, Y.; Li, L.; Bresette, W.; Chen, Y.; Cheng, J.; Lu, Y. Resveratrol exerts dose-dependent anti-fibrotic or pro-fibrotic effects in kidneys: A potential risk to individuals with impaired kidney function. Phytomedicine 2019, 57, 223–235. [Google Scholar] [CrossRef]
- Nathan, J.; Raghunathan, M. Resveratrol suppresses angiogenesis by down-regulating Vegf/Vegfr2 in Zebrafish (Danio rerio) embryos. J. Chem. Pharm. Res. 2014, 6, 892–899. [Google Scholar]
- Cavalcante, A.K.; Lopes-Ferreira; Rogero, S.O.; Rogero, J.R. Evaluation of resveratrol toxicity in the embryolarval stage of Danio rerio fish. Ecotoxicol. Environ. 2017, 12, 133–139. [Google Scholar] [CrossRef]
- Cottart, C.H.; Nivet-Antoine, V.; Laguillier-Morizot, C.; Beaudeux, J.L. Resveratrol bioavailability and toxicity in humans. Mol. Nutr. Food Res. 2010, 54, 7–16. [Google Scholar] [CrossRef]
- Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef]
- Lecumberri, E.; Dupertuis, Y.M.; Miralbell, R.; Pichard, C. Green tea polyphenol epigallocatechin-3-gallate (EGCG) as adjuvant in cancer therapy. Clin. Nutr. 2013, 32, 894–903. [Google Scholar] [CrossRef]
- Fujiki, H.; Imai, K.; Nakachi, K.; Sueoka, E.; Watanabe, T.; Suganuma, M. Innovative strategy of cancer treatment with the combination of green tea catechins and anticancer compounds. Cancer Cell Microenviron. 2015, 2. [Google Scholar]
- Sharifi-Rad, M.; Pezzani, R.; Redaelli, M.; Zorzan, M.; Imran, M.; Ahmed Khalil, A.; Salehi, B.; Sharopov, F.; Cho, W.C.; Sharifi-Rad, J. Preclinical pharmacological activities of epigallocatechin-3-gallate in signaling pathways: An update on cancer. Molecules 2020, 25, 467. [Google Scholar] [CrossRef]
- Steinmann, J.; Buer, J.; Pietschmann, T.; Steinmann, E. Anti-infective properties of epigallocatechin-3-gallate (EGCG), a component of green tea. Br. J. Pharmacol. 2013, 168, 1059–1073. [Google Scholar] [CrossRef]
- Tachibana, H. Green tea polyphenol sensing. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2011, 87, 66–80. [Google Scholar] [CrossRef] [PubMed]
- Gu, J.W.; Makey, K.L.; Tucker, K.B.; Chinchar, E.; Mao, X.; Pei, I.; Thomas, E.Y.; Miele, L. EGCG, a major green tea catechin suppresses breast tumor angiogenesis and growth via inhibiting the activation of HIF-1α and NFκB, and VEGF expression. Vasc. Cell 2013, 5, 9. [Google Scholar] [CrossRef]
- Zhou, J.; Farah, B.L.; Sinha, R.A.; Wu, Y.; Singh, B.K.; Bay, B.H.; Yang, C.S.; Yen, P.M. Epigallocatechin-3-gallate (EGCG), a green tea polyphenol, stimulates hepatic autophagy and lipid clearance. PLoS ONE 2014, 9, e87161. [Google Scholar] [CrossRef]
- Zhao, T.; Li, C.; Wang, S.; Song, X. Green Tea (Camellia sinensis): A Review of Its Phytochemistry, Pharmacology, and Toxicology. Molecules 2022, 27, 3909. [Google Scholar] [CrossRef]
- Vickery, K.D.; Sauser, K.; Davis, M.M. Policy responses to demand for health care access: From the individual to the population. JAMA 2013, 309, 665–666. [Google Scholar] [CrossRef]
- Mereles, D.; Hunstein, W. Epigallocatechin-3-gallate (EGCG) for clinical trials: More pitfalls than promises? Int. J. Mol. Sci. 2011, 12, 5592–5603. [Google Scholar] [CrossRef]
- Siriphap, A.; Kiddee, A.; Duangjai, A.; Yosboonruang, A.; Pook-In, G.; Saokaew, S.; Sutheinkul, O.; Rawangkan, A. Antimicrobial activity of the green tea polyphenol (-)-epigallocatechin-3-gallate (egcg) against clinical isolates of multidrug-resistant vibrio cholerae. Antibiotics 2022, 11, 518. [Google Scholar] [CrossRef]
- Shen, C.L.; Yeh, J.K.; Cao, J.J.; Wang, J.S. Green tea and bone metabolism. Nutr. Res. 2009, 29, 437–456. [Google Scholar] [PubMed]
- Chang, L.K.; Wei, T.T.; Chiu, Y.F.; Tung, C.P.; Chuang, J.Y.; Hung, S.K.; Li, C.; Liu, S.T. Inhibition of Epstein-Barr virus lytic cycle by (-)-epigallocatechin gallate. Biochem. Biophys. Res. Commun. 2003, 301, 1062–1068. [Google Scholar]
- Song, J.M.; Lee, K.H.; Seong, B.L. Antiviral effect of catechins in green tea on influenza virus. Antivir. Res. 2005, 68, 66–74. [Google Scholar] [CrossRef]
- Weber, J.M.; Ruzindana-Umunyana, A.; Imbeault, L.; Sircar, S. Inhibition of adenovirus infection and adenain by green tea catechins. Antivir. Res. 2003, 58, 167–173. [Google Scholar] [CrossRef]
- Legeay, S.; Rodier, M.; Fillon, L.; Faure, S.; Clere, N. Epigallocatechin gallate: A review of its beneficial properties to prevent metabolic syndrome. Nutrients 2015, 7, 5443–5468. [Google Scholar] [CrossRef]
- Khan, N.; Mukhtar, H. Tea and health: Studies in humans. Curr. Pharm. Des. 2013, 19, 6141–6147. [Google Scholar] [CrossRef]
- da Silva Pinto, M. Tea: A new perspective on health benefits. Food Res. Int. 2013, 53, 558–567. [Google Scholar] [CrossRef]
- Pang, J.Y.; Zhao, K.J.; Wang, J.B.; Ma, Z.J.; Xiao, X.H. Green tea polyphenol, epigallocatechin-3-gallate, possesses the antiviral activity necessary to fight against the hepatitis B virus replication in vitro. J. Zhejiang Univ. Sci. B 2014, 15, 533–539. [Google Scholar] [CrossRef] [PubMed]
- Goodpasture, C.E.; Arrighi, F.E. Effects of food seasonings on the cell cycle and chromosome morphology of mammalian cells in vitro with special reference to turmeric. Food Cosmet. Toxicol. 1976, 14, 9–14. [Google Scholar] [CrossRef] [PubMed]
- Giri, A.K.; Das, S.K.; Talukder, G.; Sharma, A. Sister chromatid exchange and chromosome aberrations induced by curcumin and tartrazine on mammalian cells in vivo. Cytobios 1990, 62, 111–117. [Google Scholar]
- Sakano, K.; Kawanishi, S. Metal-mediated DNA damage induced by curcumin in the presence of human cytochrome P450 isozymes. Arch. Biochem. Biophys. 2002, 405, 223–230. [Google Scholar] [CrossRef]
- Nair, J.; Strand, S.; Frank, N.; Knauft, J.; Wesch, H.; Galle, P.R.; Bartsch, H. Apoptosis and age-dependant induction of nuclear and mitochondrial etheno-DNA adducts in Long-Evans Cinnamon (LEC) rats: Enhanced DNA damage by dietary curcumin upon copper accumulation. Carcinogenesis 2005, 26, 1307–1315. [Google Scholar] [CrossRef]
- Blasiak, J.; Trzeciak, A.; Kowalik, J. Curcumin damages DNA in human gastric mucosa cells and lymphocytes. J. Environ. Pathol. Toxicol. Oncol. 1999, 18, 271–276. [Google Scholar]
- Verschoyle, R.D.; Steward, W.P.; Gescher, A.J. Putative cancer chemopreventive agents of dietary origin—How safe are they? Nutr. Cancer 2007, 59, 152–162. [Google Scholar] [CrossRef]
- National Toxicology Program. 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. [Google Scholar]
- Dance-Barnes, S.T.; Kock, N.D.; Moore, J.E.; Lin, E.Y.; Mosley, L.J.; D’Agostino, R.B., Jr.; McCoy, T.P.; Townsend, A.J.; Miller, M.S. Lung tumor promotion by curcumin. Carcinogenesis 2009, 30, 1016–1023. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.; Lu, J.; Holmgren, A. Thioredoxin reductase is irreversibly modified by curcumin: A novel molecular mechanism for its anticancer activity. J. Biol. Chem. 2005, 280, 25284–25290. [Google Scholar] [CrossRef]
- McNally, S.J.; Harrison, E.M.; Ross, J.A.; Garden, O.J.; Wigmore, S.J. Curcumin induces heme oxygenase 1 through generation of reactive oxygen species, p38 activation and phosphatase inhibition. Int. J. Mol. Med. 2007, 19, 165–173. [Google Scholar] [CrossRef]
- Atsumi, T.; Fujisawa, S.; Tonosaki, K. Relationship between intracellular ROS production and membrane mobility in curcumin- and tetrahydrocurcumin-treated human gingival fibroblasts and human submandibular gland carcinoma cells. Oral Dis. 2005, 11, 236–242. [Google Scholar] [CrossRef] [PubMed]
- Sandur, S.K.; Ichikawa, H.; Pandey, M.K.; Kunnumakkara, A.B.; Sung, B.; Sethi, G.; Aggarwal, B.B. Role of pro-oxidants and antioxidants in the anti-inflammatory and apoptotic effects of curcumin (diferuloylmethane). Free Radic. Biol. Med. 2007, 43, 568–580. [Google Scholar] [CrossRef]
- Moos, P.J.; Edes, K.; Mullally, J.E.; Fitzpatrick, F.A. Curcumin impairs tumor suppressor p53 function in colon cancer cells. Carcinogenesis 2004, 25, 1611–1617. [Google Scholar] [CrossRef]
- Lopez-Lazaro, 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–S127. [Google Scholar] [PubMed]
- Vareed, S.K.; Kakarala, M.; Ruffin, M.T.; Crowell, J.A.; Normolle, D.P.; Djuric, Z.; Brenner, D.E. Pharmacokinetics of curcumin conjugate metabolites in healthy human subjects. Cancer Epidemiol. Biomarkers Prev. 2008, 17, 1411–1417. [Google Scholar] [CrossRef] [PubMed]
- Ireson, C.; Orr, S.; Jones, D.J.; Verschoyle, R.; Lim, C.K.; Luo, J.L.; Howells, L.; Plummer, S.; Jukes, R.; Williams, M.; 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, 1058–1064. [Google Scholar]
- Ireson, C.R.; Jones, D.J.; Orr, S.; Coughtrie, M.W.; Boocock, D.J.; Williams, M.L.; Farmer, P.B.; Steward, W.P.; Gescher, A.J. Metabolism of the cancer chemopreventive agent curcumin in human and rat intestine. Cancer Epidemiol. Biomarkers Prev. 2002, 11, 105–111. [Google Scholar] [PubMed]
- Mancuso, C.; Barone, E. Curcumin in clinical practice: Myth or reality? Trends Pharmacol. Sci. 2009, 30, 333–334. [Google Scholar] [CrossRef]
- Oetari, S.; Sudibyo, M.; Commandeur, J.N.; Samhoedi, R.; Vermeulen, N.P. Effects of curcumin on cytochrome P450 and glutathione S-transferase activities in rat liver. Biochem. Pharmacol. 1996, 51, 39–45. [Google Scholar] [CrossRef] [PubMed]
- Appiah-Opong, R.; Commandeur, J.N.; Vugt-Lussenburg, B.; Vermeulen, N.P. Inhibition of human recombinant cytochrome P450s by curcumin and curcumin decomposition products. Toxicology 2007, 235, 83–91. [Google Scholar] [CrossRef] [PubMed]
- Thapliyal, R.; Maru, G.B. Inhibition of cytochrome P450 isozymes by curcumins in vitro and in vivo. Food Chem. Toxicol. 2001, 39, 541–547. [Google Scholar] [CrossRef] [PubMed]
- Jiao, Y.; Wilkinson, J.; Di, X.; Wang, W.; Hatcher, H.; Kock, N.D.; D’Agostino, R., Jr.; Knovich, M.A.; Torti, F.M.; Torti, S.V. Curcumin, a cancer chemopreventive and chemotherapeutic agent, is a biologically active iron chelator. Blood 2009, 113, 462–469. [Google Scholar] [CrossRef] [PubMed]
- Means, R.T., Jr. Ironing out complementary medicine. Blood 2009, 113, 270–271. [Google Scholar] [CrossRef] [PubMed]
- Kocaadam, B.; Şanlier, N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef]
- Sharma, R.A.; Euden, S.A.; Platton, S.L.; Cooke, D.N.; Shafayat, A.; Hewitt, H.R.; Marczylo, T.H.; Morgan, B.; Hemingway, D.; Plummer, S.M. Phase I clinical trial of oral curcumin: Biomarkers of systemic activity and compliance. Clin. Cancer Res. 2004, 10, 6847–6854. [Google Scholar] [CrossRef]
- Gubin, M.M.; Vesely, M.D. Cancer Immunoediting in the Era of Immuno-oncology. Clin. Cancer Res. 2020, 28, 3917–3928. [Google Scholar]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sochacka, K.; Lachowicz-Wiśniewska, S. Phytotherapy and the Role of Bioactive Compounds in Modulating Mechanisms of Overweight and Obesity Comorbid with Depressive Symptoms—A Scoping Review of Mechanisms of Action. Molecules 2025, 30, 2827. https://doi.org/10.3390/molecules30132827
Sochacka K, Lachowicz-Wiśniewska S. Phytotherapy and the Role of Bioactive Compounds in Modulating Mechanisms of Overweight and Obesity Comorbid with Depressive Symptoms—A Scoping Review of Mechanisms of Action. Molecules. 2025; 30(13):2827. https://doi.org/10.3390/molecules30132827
Chicago/Turabian StyleSochacka, Klaudia, and Sabina Lachowicz-Wiśniewska. 2025. "Phytotherapy and the Role of Bioactive Compounds in Modulating Mechanisms of Overweight and Obesity Comorbid with Depressive Symptoms—A Scoping Review of Mechanisms of Action" Molecules 30, no. 13: 2827. https://doi.org/10.3390/molecules30132827
APA StyleSochacka, K., & Lachowicz-Wiśniewska, S. (2025). Phytotherapy and the Role of Bioactive Compounds in Modulating Mechanisms of Overweight and Obesity Comorbid with Depressive Symptoms—A Scoping Review of Mechanisms of Action. Molecules, 30(13), 2827. https://doi.org/10.3390/molecules30132827