Next Article in Journal
Structural and Quantitative Analysis of Polyfluoroalkyl Substances (PFASs) and Para-Phenylenediamines (PPDs) by Direct Analysis in Real Time Ion Mobility Mass Spectrometry (DART-IM-MS)
Previous Article in Journal
Ferrocenyl Substituted Stannanethione and Stannaneselone
Previous Article in Special Issue
Cosmeceutical and Wound-Healing Activities of Green Hydroxypropyl-β-Cyclodextrin-Glycerol-Based Satureja montana Extracts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

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

by
Klaudia Sochacka
and
Sabina Lachowicz-Wiśniewska
*
Faculty of Medicine and Health Science, University of Kalisz, pl. W. Bogusławskiego 2, 62-800 Kalisz, Poland
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(13), 2827; https://doi.org/10.3390/molecules30132827
Submission received: 1 June 2025 / Revised: 24 June 2025 / Accepted: 25 June 2025 / Published: 30 June 2025
(This article belongs to the Special Issue Bioactive Compounds from Foods for Health Benefits)

Abstract

Obesity and depression frequently coexist, sharing overlapping molecular pathways such as inflammation, oxidative stress, gut microbiota dysbiosis, and neuroendocrine dysfunction. Recent research highlights the therapeutic potential of plant-derived bioactive compounds in targeting these shared mechanisms. This scoping review followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and included 261 peer-reviewed studies identified through PubMed, Scopus, and the Web of Science up to December 2024. Studies were screened based on predefined inclusion and exclusion criteria. This review synthesizes data from peer-reviewed studies, including both preclinical and clinical investigations, focusing on polyphenols, flavonoids, alkaloids, and other phytochemicals with anti-inflammatory, antioxidant, neuroprotective, and metabolic effects. Compounds such as quercetin, epigallocatechin gallate (EGCG), resveratrol, curcumin, anthocyanins, and luteolin demonstrate promise in modulating adenosine monophosphate-activated protein kinase (AMPK), brain-derived neurotrophic factor (BDNF), nuclear factor kappa B (NF-κB), and gut–brain axis pathways. Our scoping review, conducted in accordance with PRISMA guidelines, identifies promising combinations and mechanisms for integrative phytotherapy. These findings underscore the potential of botanical strategies in developing future interventions for metabolic and mood comorbidities.

Graphical Abstract

1. Introduction

Obesity remains one of the most critical and escalating global public health concerns. Despite extensive research efforts, effective treatment strategies are still limited, and the global prevalence of obesity continues to rise at an alarming rate [1]. Obesity is defined as the excessive accumulation of adipose tissue resulting from a sustained positive energy balance, which contributes to the development of various metabolic and/or inflammatory disorders [2]. This condition poses a substantial health burden on modern populations. A significant complication is its frequent coexistence with depression, a condition often referred to as a “silent pandemic” due to its complex social and health consequences. Both conditions are associated with high morbidity and mortality, affecting millions of individuals globally.
Depression is the most common mental illness, characterized by symptoms such as persistent low mood, anhedonia, feelings of guilt, impaired concentration, low self-esteem, sleep disturbances, and appetite dysregulation—either hyperphagia or hypophagia [3]. According to a meta-analysis by Luppino et al. [4], a bidirectional association exists between obesity and depression: each condition increases the risk of developing the other. Obesity may predispose individuals to depression, whereas depression can act as a risk factor for the onset of obesity [4].
In the search for new therapeutic strategies to address both obesity and depression, increasing attention has been directed toward plant-derived materials. Owing to their high content of bioactive phytochemicals, plant-derived substances have emerged as promising adjunctive agents in the treatment of both conditions. Research indicates that certain plant-based compounds may mitigate chronic inflammation, thereby supporting patient health. Among the most promising bioactive compounds with anti-obesity potential are polyphenols (such as resveratrol, quercetin, and epigallocatechin gallate), betalains, omega-3 fatty acids, and dietary fiber [5].
Depression is a condition in which inflammatory processes can significantly exacerbate clinical symptoms. Inflammation may represent not only a physiological manifestation of depression but also a potential etiological factor [6]. Therefore, the anti-inflammatory and antioxidant properties of natural bioactive compounds, as with obesity, are of considerable importance in the supportive management of depression. Several plant-derived compounds have demonstrated antidepressant-like effects in preclinical and clinical models, including apigenin, berberine, curcumin, genistein, luteolin, N-acetylcysteine, naringenin, piperine, quercetin, resveratrol, epigallocatechin gallate, and baicalin [7].
This review aims to critically evaluate the current scientific evidence on the health-promoting potential of selected plant-derived bioactive compounds in the prevention and adjunctive treatment of obesity and depression. Emphasis is placed on molecular and cellular mechanisms, including anti-inflammatory, antioxidant, neuroprotective, and metabolic pathways. Both in vitro and in vivo studies are included to provide a comprehensive overview of the dual-action phytotherapeutic approach.

2. Materials and Methods

This review was conducted following PRISMA guidelines for scoping reviews. A structured literature search was performed using PubMed, Scopus, Web of Science, and Google Scholar databases for publications published between January 2000 and December 2024 (Figure 1). The search strategy included the following keywords and their combinations: “obesity and depression,” “bioactive compounds,” “polyphenols and inflammation,” “curcumin and neuroprotection,” “EGCG and metabolism,” and “phytotherapy and mood disorders.” Boolean operators were used to refine the search, and Medical Subject Headings (MeSH) terms were applied where appropriate.
The final search was conducted on 20 December 2024. The search results were imported into EndNote for duplicate removal, and studies were screened based on titles and abstracts, followed by full-text evaluation. Two independent reviewers conducted the selection process. Discrepancies were resolved through discussion or, if needed, consultation with a third reviewer.
Inclusion and exclusion criteria were applied as follows: (i) included: peer-reviewed articles in English focusing on plant-derived bioactive compounds with mechanistic or interventional relevance to obesity and/or depression, including both preclinical (in vitro and in vivo) and clinical studies; (ii) excluded: non-peer-reviewed articles, review articles, case reports, editorials, conference abstracts, studies without mechanistic data, and duplicates.
A total of 261 references were ultimately included and analyzed. Risk of bias was not formally assessed due to the exploratory nature of the scoping review. This review was not registered in PROSPERO or any other platform.
Data extraction was performed using a standardized form that included study type (in vitro, in vivo, clinical), bioactive compound (e.g., curcumin, EGCG, resveratrol), mechanism of action (e.g., modulation of AMPK, BDNF, NF-κB), health outcome (e.g., anti-inflammatory, antioxidant, antidepressant, anti-obesity), and other relevant parameters. Extracted data were synthesized qualitatively.
The final pool of articles (n = 261) was analyzed narratively with particular attention to biochemical pathways, pharmacological targets, and the potential synergistic effects of bioactive compounds in the context of obesity and depression comorbidity. Among the included studies, approximately 45 were in vitro studies, 70 were conducted in vivo using animal models, and 55 were clinical investigations (observational and interventional). The most frequently studied phytochemicals were polyphenols, including quercetin, resveratrol, epigallocatechin gallate (EGCG), curcumin, anthocyanins, and luteolin. These compounds were shown to exert antioxidant, anti-inflammatory, neuroprotective, and metabolic effects, often through the modulation of AMPK, NF-κB, BDNF, the hypothalamic–pituitary–adrenal (HPA) axis, and gut microbiota. Approximately 100 studies addressed anti-inflammatory effects, 60 explored antioxidant mechanisms, 40 focused on neuroprotective properties, and 61 examined anti-obesity effects. The reviewed data highlight the multidimensional potential of plant-derived bioactive compounds in targeting interrelated pathophysiological pathways in metabolic and mood disorders.

3. Epidemiological and Public Health Impact of Obesity and Depression

According to the Global Burden of Disease study [1], obesity and depression rank among the most significant global health challenges. Their co-occurrence poses a serious threat to patient health and well-being [8]. Both obesity and depression are often described as epidemics of the 21st century and constitute significant public health concerns, particularly due to the escalating costs associated with their treatment. In recent years, greater attention has been directed toward the psychological implications of obesity, including low self-esteem, body image dissatisfaction, and social withdrawal. These psychosocial consequences frequently contribute to chronic stress, emotional dysregulation, and, ultimately, the onset of depressive symptoms. These psychosocial issues may lead to chronic stress, a persistently low mood, and ultimately, the development of depression.
Conversely, depression—which is characterized by persistent sadness, anhedonia, and stress vulnerability—may promote maladaptive eating behaviors such as emotional overeating or food restriction. These behaviors often serve as coping mechanisms for psychological distress and can lead to or exacerbate obesity [9]. Globally, the prevalence of both conditions continues to escalate. According to the World Health Organization (WHO), the global prevalence of obesity has nearly tripled since 1975, while over 300 million individuals are currently affected by mental health disorders, with depression being one of the most commonly diagnosed psychiatric conditions [10].
In the Polish context, national surveillance data reveal a disturbing upward trend in obesity rates, particularly among children and adolescents, positioning Poland among the leading European countries in this regard [11]. The growing incidence of both obesity and depression contributes significantly to the burden of non-communicable diseases (NCDs), especially in high-income and rapidly developing nations. The treatment and management of patients suffering from comorbid obesity and depression generate considerable strain on healthcare resources. These burdens include extended treatment durations, frequent relapses, reduced treatment efficacy, and increased incidence of comorbid complications, such as cardiovascular disease, type 2 diabetes, and anxiety disorders, further exacerbating public health and economic challenges [12,13].

4. The Role of Major Classes of Bioactive Compounds in the Treatment of Obesity and Depression

Plant-derived bioactive compounds—including polyphenols, phenolic acids, polysaccharides, saponins, omega-3 fatty acids, and probiotics—have shown therapeutic potential in obesity and depression due to their ability to modulate inflammation, oxidative stress, lipid metabolism, and the gut–brain axis. Their mechanisms involve the inhibition of adipogenesis, the regulation of transcription factors (e.g., PPARγ and C/EBPα), the suppression of pro-inflammatory cytokines (e.g., TNF-α and IL-6) via NF-κB inhibition, and neurochemical modulation [14,15,16,17,18,19].
Polyphenols are the most abundant phytochemicals in the human diet and are recognized for their anti-obesity and neuroprotective effects. Compounds such as quercetin, resveratrol, curcumin, and catechins inhibit adipogenesis and lipogenesis, stimulate fatty acid oxidation, and promote adipocyte apoptosis [14,15,16,17,18,19]. Hsu and Yen [20] demonstrated that rutin significantly reduces intracellular triglycerides and downregulates key adipogenic factors while increasing adiponectin levels, thereby supporting improved lipid profiles. Additionally, polyphenols enhance antioxidant enzyme activity and reduce oxidative stress associated with adiposity and neuroinflammation [20].
Phenolic acids also exert strong antioxidant and anti-inflammatory properties. In vitro studies show that they suppress macrophage infiltration and reduce cytokines such as MCP-1 and PAI-1 via NF-κB inhibition, while modulating adipokine secretion [21,22,23]. Their ability to maintain a redox balance makes them valuable agents in preventing chronic metabolic and mood disorders.
Polysaccharides are complex carbohydrates with immunomodulatory and anti-inflammatory properties. Their consumption influences the regulation of the gut microbiota, which is crucial for both metabolic and mental health. Polysaccharides can increase satiety, which promotes weight control, while at the same time they induce beneficial changes in the nervous system, alleviating symptoms of depression by affecting the brain-gut axis and modulating neurotransmitters [24,25,26,27,28,29,30,31,32,33]. Zou et al. [34] reported that insoluble fiber enhances IL-22 expression, contributing to intestinal barrier integrity and microbial defense. In contrast, a study by Sanchez et al. [35] indicates that soluble fiber intake may increase obesity-associated inflammation because the fiber helps protect tissues from the oxidative stress characteristic of the disease [36]. Moreover, studies by Ma et al. [36] confirmed that both soluble and insoluble fibers reduce CRP and LPS levels, thus lowering systemic inflammation and improving metabolic parameters [34,35,36,37,38].
Omega-3 fatty acids (such as EPA and DHA) contribute to the resolution of inflammation through the activation of PPARγ and the inhibition of NF-κB, which reduces pro-inflammatory cytokine levels and improves insulin sensitivity. Probiotic strains, particularly Lactobacillus and Bifidobacterium, enhance gut microbial balance, reduce intestinal permeability, and decrease LPS-induced inflammation. Their synergistic effect with bioactive plant compounds further supports their use in the management of metabolic and neuropsychiatric conditions [39,40].
In summary, different classes of natural bioactive compounds modulate shared pathophysiological pathways involved in obesity and depression. Through combined anti-inflammatory, antioxidant, and microbiota-regulating actions, they hold promise as adjunct therapeutic agents. While oral supplementation remains the most common route of administration, further clinical trials are needed to confirm long-term efficacy, optimize dosage, and evaluate safety.

5. The Role of Bioactive Compounds in the Management of Obesity and Depression

Although this review primarily focuses on the bioactivity and mechanisms of plant-derived compounds, it is important to note that phytotherapy may be administered via various routes, each potentially influencing efficacy, particularly in mood disorders. While oral delivery (e.g., capsules, teas, tinctures) remains the most common and practical approach, it is often subject to first-pass hepatic metabolism and variable absorption.
Alternative routes—such as inhalation (e.g., aromatherapy with lavender or bergamot oils), transdermal application, or sublingual tinctures—may offer faster onset and bypass hepatic degradation. Intraperitoneal and intravenous administration have also been explored in experimental models to isolate specific mechanistic effects [41,42,43,44].
Most in vivo and clinical studies reviewed here employed oral administration due to its ease and regulatory acceptance. However, future research should evaluate the pharmacokinetics and comparative efficacy of novel formulations, such as nanoemulsions (e.g., nanoencapsulated curcumin), liposomes, and transdermal systems, to improve therapeutic outcomes and patient adherence [45,46,47,48,49].

The Role and Classification of Bioactive Compounds in Therapy

Nature offers an almost limitless reservoir of compounds with potential health benefits, commonly referred to as natural bioactive substances. Bioactive compounds are naturally occurring substances that can exert favorable effects on human health. In the context of nutrition, they have been described as nutraceuticals since 1979, as their consumption provides health benefits that extend beyond basic nutritional needs [50]. These valuable substances can be classified into four main categories: macronutrients, micronutrients, phytochemicals, and regulators of the gut microbiota [51].
Macronutrients include carbohydrates, fats, and proteins. Micronutrients comprise essential vitamins and minerals that are indispensable for proper physiological functioning. Phytochemicals include diverse groups such as terpenes, alkaloids, and phenolic compounds. Lastly, the microbiota-regulating group includes probiotics, prebiotics, synbiotics, and postbiotics [52].
This review specifically focuses on phenolic compounds—especially polyphenols—due to their demonstrated efficacy in targeting inflammation, oxidative stress, metabolic dysregulation, and neurochemical imbalances that are commonly shared between obesity and depression (Figure 2).

6. Selected Bioactive Compounds and Their Mechanisms of Action

Obesity and depression share overlapping inflammatory, metabolic, and neuroendocrine pathways. Chronic low-grade inflammation, which is characterized by the increased secretion of pro-inflammatory cytokines (TNF-α, IL-6, and MCP-1), dysregulated adipokine profiles (leptin, resistin, and adiponectin), and oxidative stress, contributes to both conditions. Several plant-derived compounds, particularly polyphenols, modulate key molecular targets, such as AMPK, NF-κB, PPARγ, and BDNF, thereby improving metabolic homeostasis and CNS function. Below, selected phytochemicals are discussed with emphasis on their dual activity.

6.1. The Treatment of Obesity and Its Mechanisms of Action

Excessive adipose tissue expansion in obesity promotes chronic low-grade inflammation, largely through the dysregulated secretion of adipokines and pro-inflammatory cytokines, such as TNF-α, IL-6, MCP-1, resistin, and visfatin [53,54,55,56]. This inflammatory milieu, exacerbated by the increased production of ROS and RNS, contributes to metabolic disturbances and comorbidities like type 2 diabetes and cardiovascular disease [57] (Table 1). Bioactive compounds may counteract these effects by modulating key molecular pathways, including PPARα, COX-2, GLUT4, SIRT1, and PGC1-α, thereby improving energy metabolism and inflammatory balance [5,58].
Polyphenols are prominent dietary phytochemicals that are found in fruits, vegetables, tea, and whole grains [59,60,61,62]. They include flavonoids, phenolic acids, stilbenes, and curcuminoids, and exhibit anti-obesity activity by suppressing adipogenesis and lipogenesis, inducing adipocyte apoptosis, and downregulating lipogenic genes such as PPARγ and SREBP1 [63,64,65]. Polyphenols also support gut microbiota modulation and reduce inflammatory cytokine expression, contributing to systemic metabolic regulation [66]. Compounds such as quercetin, resveratrol, EGCG, and genistein primarily inhibit adipocyte differentiation, positioning them as promising agents for obesity prevention and body weight control [67].
Quercetin, a flavonoid that is abundant in onions, garlic, apples, and citrus fruits, is recognized as one of the most potent plant-derived antioxidants. Although numerous in vitro and in vivo studies confirm its anti-obesity potential, human clinical data remain limited [67,68,69]. In vitro, quercetin inhibits adipogenesis in preadipocytes and reduces inflammatory cytokine secretion in macrophages and adipocytes exposed to TNF-α [70,71,72,73]. It also improves insulin sensitivity in inflamed adipocytes [72,74,75].
A clinical trial using 150 mg/day quercetin supplementation in overweight individuals with different ApoE genotypes demonstrated reduced waist circumference and triglyceride levels [76]. In vivo, quercetin inhibited adipogenic marker expression in F344 rat muscle progenitor cells [77]. Beyond metabolic benefits, it also exhibits antidepressant-like effects by lowering plasma cortisol, improving memory, and reducing anxiety- and depressive-like behavior [78,79,80], potentially through the modulation of BDNF and iNOS.
Genistein, an isoflavone found in soy, has demonstrated anti-obesity activity through the modulation of lipid metabolism and energy balance [81]. Mice supplemented with 600 mg or 1500 mg/kg genistein showed significant weight loss, improved glucose and lipid profiles, and enhanced adipocyte apoptosis [82,83]. Additionally, 0.2% genistein included in a high-fat diet improved glucose uptake and reduced hepatic steatosis [84]. These effects are linked to increased lipolysis, fatty acid oxidation, and the upregulation of lipid metabolism genes [85].
Emerging studies have suggested that genistein may also exert antidepressant-like effects, possibly via serotonergic 5-HT1A receptor interaction and the inhibition of monoamine oxidase (MAO), which enhances brain serotonin, dopamine, and norepinephrine levels [86,87,88,89]. In vivo, genistein’s efficacy was comparable to amitriptyline at 10 mg/kg [88]. Given these findings, genistein is a promising nutraceutical candidate—e.g., standardized soy isoflavone supplements (30–50 mg/dose)—with the potential for bioavailability-enhanced formulations. Future clinical trials should target overweight adults with insulin resistance and assess both metabolic and inflammatory endpoints.
Resveratrol, a polyphenol that is highly concentrated in red grape skins, is also found in tea, berries, pomegranate, nuts, and dark chocolate [90]. It possesses strong anti-inflammatory and antioxidant properties and is known to activate sirtuin 1 (SIRT1), a regulator of mitochondrial function and cellular metabolism [91]. Resveratrol also modulates AMP-activated protein kinase (AMPK), thereby promoting energy balance and lipid homeostasis.
Through AMPK activation, resveratrol inhibits adipocyte differentiation and lipogenesis while promoting lipolysis and β-oxidation [92]. In vitro, it enhances apoptosis in mature adipocytes and reduces fat accumulation [93,94,95]. In vivo rodent studies confirm reductions in fat mass via the suppression of lipid deposition and the stimulation of oxidative pathways [96,97].
Beyond metabolic effects, resveratrol exhibits neuroprotective activity. It elevates dopamine and serotonin levels in the prefrontal cortex and upregulates neuropeptide Y (NPY), thereby influencing mood, appetite, and stress responses [98,99]. Studies in corticosterone-induced depression models demonstrate that resveratrol improves mood by modulating the HPA axis and increasing hippocampal BDNF levels [100]. Additional evidence suggests that it promotes neurogenesis, regulates SIRT1, and suppresses NF-κB signaling, collectively contributing to its antidepressant effects [101].
Epigallocatechin gallate (EGCG), the primary catechin in green tea, exerts potent anti-obesity effects by influencing both nutrient absorption and intracellular energy regulation. One of its main molecular targets is AMP-activated protein kinase (AMPK), which is a central regulator of cellular energy homeostasis. EGCG activates AMPK in the liver, skeletal muscle, and adipose tissue by increasing the AMP/ATP ratio, thereby promoting fatty acid oxidation and inhibiting lipogenesis [102].
AMPK activation by EGCG results in the phosphorylation of acetyl-CoA carboxylase (ACC), the inhibition of lipogenic enzymes (acc1, fas, scd1), and the downregulation of transcription factors, such as C/EBPβ, PPARγ, and SREBP1 [102,103,104]. Simultaneously, it enhances lipolysis by upregulating hormone-sensitive lipase (HSL) and related enzymes, and promotes mitochondrial biogenesis via PGC-1α expression, contributing to increased energy expenditure [103].
EGCG also exhibits anti-inflammatory properties by suppressing IL-1β and TNF-α production through the inhibition of the NF-κB and MAPK pathways [105]. Its neuroprotective effects include the modulation of the HPA axis, the regulation of neurosteroid biosynthesis, and the protection of hippocampal DNA from oxidative and apoptotic damage [106].
In a study by Li et al. [107], HFD-fed mice treated with EGCG (50–100 mg/kg/day, 20 weeks) showed significant reductions in body and epididymal fat mass, improved lipid profiles (including lower TG, cholesterol, HDL-C, and LDL-C), and increased fecal excretion of fatty acids. Collectively, EGCG promotes weight loss and metabolic balance through AMPK activation, lipogenesis inhibition, thermogenesis stimulation, and anti-inflammatory and neuroprotective actions [103,105,106,107,108,109].
Anthocyanins, a subclass of phenolic compounds found in purple sweet potatoes, black soybeans, berries, and red fruits, play a central role in regulating energy metabolism and reducing obesity risk. Their key mechanism involves the activation of AMP-activated protein kinase (AMPK), which promotes catabolic processes such as fatty acid oxidation and inhibits lipogenesis and cholesterol synthesis [104,105,106]. Hwang et al. [108] showed that anthocyanins from purple sweet potato activated AMPK in HepG2 cells, thereby reducing hepatic lipid accumulation and body weight in obese mice (200 mg/kg/day). Similar effects were observed with black soybean extract containing cyanidin-3-glucoside [109].
AMPK activation also enhances mitochondrial biogenesis via PGC-1α and upregulates PPARγ, thereby increasing mitochondrial function and reducing adipogenesis [110,111]. Anthocyanins inhibit pancreatic lipase activity, thereby decreasing fat absorption and serum free fatty acids, as shown with lychee flower extract and pomegranate or red orange juices [112]. They also modulate glucose metabolism by inhibiting α-glucosidase and enhancing insulin sensitivity in muscle and liver tissues through AMPK signaling [113].
Moreover, anthocyanins regulate appetite-related hormones. They reduce leptin levels and affect the secretion of ghrelin, CCK, GIP, and GLP-1, thereby enhancing satiety and reducing caloric intake, particularly in individuals with insulin resistance [114,115]. Their prebiotic effects include the stimulation of beneficial microbiota (including Lactobacillus, Bifidobacterium, and Enterococcus), leading to increased short-chain fatty acid production, which further modulates inflammation, appetite, and insulin response [108,109,116,117].
Betalains, which are found in beetroot (Beta vulgaris) and dragon fruit (Hylocereus spp.), are bioactive pigments with potent antioxidant and anti-inflammatory properties. Betanin, the primary red-violet pigment in beets, reduces ROS, RNS, and proinflammatory cytokines (e.g., TNF-α, IL-1β, and IL-6), as demonstrated by Ahmadi et al. [118]. In a NASH mouse model, Yahaghi et al. [119] found that 20 mg/kg of intraperitoneal betalain administration increased adiponectin and decreased leptin levels, although other studies reported limited effects on lipid profile or body weight [120].
In vitro assays confirm betalains’ ROS-scavenging potential using FRAP, ORAC, and ABTS methods. They also inhibit LOX-1, an enzyme involved in ROS generation [121,122,123]. Betalains isolated from quinoa (Chenopodium quinoa) further support their antioxidant efficacy, suggesting potential in mitigating oxidative stress-related metabolic disorders (Figure 3) [123,124].

6.2. Treatments for Depression Comorbid with Obesity and Their Mechanisms of Action

Building on the general mechanisms introduced earlier, a range of specific plant-derived bioactive compounds has demonstrated therapeutic effects in the context of depression coexisting with obesity. These compounds modulate neurotransmission, attenuate oxidative stress, and influence the hypothalamic–pituitary–adrenal (HPA) axis, thereby contributing to neuroprotection and mood regulation [125,126,127,128,129,130]. Their antidepressant effects are attributed to interactions with monoaminergic systems, the normalization of neurotransmitter levels, and the modulation of inflammatory and neurotrophic signaling pathways (Figure 4 and Figure 5) [131,132].
Apigenin, a flavonoid that is abundant in citrus fruits, exerts antidepressant effects through multiple mechanisms, including the inhibition of inflammatory mediators (iNOS, COX-2), the downregulation of NF-κB signaling, and the activation of neurotrophic pathways, such as p38/MAPK and PI3K/Akt [133,134,135,136,137]. Animal studies have shown increased hippocampal BDNF expression following apigenin administration, highlighting its neuroplasticity-enhancing potential.
Naringenin, another citrus-derived flavonoid, modulates serotonin and norepinephrine levels and restores HPA axis homeostasis. It reduces serum corticosterone, enhances glutathione reductase activity, and decreases pro-inflammatory cytokines (such as IL-1β, IL-6, and TNF-α), while increasing BDNF levels in the hippocampus. Naringenin’s dual modulation of mood and metabolic parameters makes it a promising candidate for mood-support functional beverages (e.g., citrus-based drinks enriched with nanoencapsulated naringenin or citrus peel extracts). Clinical trials could evaluate its synergistic effects when co-administered with selective probiotics or antidepressant therapy in patients with metabolic syndrome and depressive symptoms [138,139,140,141,142,143,144].
Curcumin, the principal curcuminoid from Curcuma longa, has been shown to inhibit monoamine oxidase (MAO-A and MAO-B), increase monoamine neurotransmitter levels, and suppress pro-inflammatory signaling (notably via NF-κB). These mechanisms jointly contribute to its antidepressant efficacy demonstrated in both preclinical and clinical models [145,146,147,148,149,150,151,152].
Piperine, the main alkaloid in black pepper, also inhibits MAO enzymes, elevates central monoamine levels, and regulates HPA axis overactivity. Its antidepressant action is further supported by increased BDNF expression and the suppression of oxidative and inflammatory pathways [153,154,155,156,157,158].
Luteolin, which is found in celery, green pepper, and Perilla, crosses the blood–brain barrier and acts as an antioxidant and cytokine inhibitor. It attenuates NF-κB and TLR4 signaling, inhibits monoamine reuptake, and normalizes corticosterone levels, contributing to improved mood and neuroprotection in chronic stress models [159,160,161,162,163,164].
Baicalin, a flavone glycoside with anti-inflammatory and antioxidant properties, activates key intracellular signaling cascades (e.g., AMPK and PI3K/Akt) and enhances neuroplasticity via the TrkB, synaptophysin, and Rac1 pathways [165]. Preclinical studies have shown that baicalin ameliorates depressive-like behaviors in chronic stress models. A meta-analysis of 22 animal studies confirmed its efficacy in standard behavioral paradigms (SPT, TST, FST, and OFT), particularly at doses between 40 and 100 mg/kg [166,167,168,169]. These results point toward baicalin and other flavonoids, such as apigenin or luteolin, as promising candidates for future diet-based interventions or adjunctive nutraceutical therapies. A translational approach may involve testing standardized plant extracts in phase I/II trials with validated psychiatric scales (e.g., HDRS and BDI-II) and inflammatory markers (e.g., IL-6 and CRP) to assess mood stabilization in obese individuals with mild to moderate depressive symptoms [170].
These findings underline the multifaceted roles of bioactive plant compounds in addressing depression co-occurring with obesity through their synergistic anti-inflammatory, antioxidant, neurotrophic, and endocrine-modulating actions (Figure 4, Table 1).
Table 1. Key bioactive compounds and their mechanisms.
Table 1. Key bioactive compounds and their mechanisms.
CompoundNatural SourceMechanism of ActionReferences
QuercetinOnions, apples, and berriesAntioxidant, anti-inflammatory, modulates AMPK and cytokines[67,68,69,70,71,72,73,74,75,76,77,78,79,80]
ApigeninParsley, celery, and chamomileAnxiolytic, neuroprotective, regulates BDNF, mTOR/AMPK/ULK1[133,134,135,136,137]
LuteolinCelery, green pepper, and chamomileReduces neuroinflammation, affects ER stress, and monoamine transporters[159,160,161,162,163,164]
NaringeninCitrus fruitsAntioxidant, modulates BDNF signaling, antidepressant[138,139,140,141,142,143,144]
BaicalinScutellaria baicalensisAnti-inflammatory, modulates PI3K/Akt, CREB/BDNF, and BBB protection[165,166,167,168,169,170]
GenisteinSoy products, soybeansPhytoestrogenic, affects neurotransmitter systems, regulates lipid metabolism[81,82,83,84,85,86,87,88,89]
ResveratrolGrapes, red wine, berriesActivates SIRT1 and AMPK, anti-adipogenic, neuroprotective[90,91,92,93,94,95,96,97,98,99,100,101]
CurcuminTurmericAnti-inflammatory, modulates NF-κB, serotonin, and dopamine[145,146,147,148,149,150,151,152]
EGCGGreen teaModulates metabolism, anti-obesity, and neuroprotective, insulin sensitivity[102,103,104,105,106,107]
AnthocyaninsBerries, red cabbage, purple cornAnti-inflammatory, modulate adipogenesis and lipid metabolism, antioxidant, influence gut-brain axis[102,103,104,105,106,107]
BetalainsBeetroot, pitayaAntioxidant, anti-inflammatory, inhibits peroxynitrite[118,119,120,121,122,123,124]
PiperineBlack pepperEnhances bioavailability, MAO inhibition, neuroactive[153,154,155,156,157,158]
Omega-3 fatty acidsFish oil, flaxseedAnti-inflammatory, neuroprotective[170,171,172,173,174,175,176,177,178]

7. In Vitro and In Vivo Studies on Bioactive Compounds

Recent studies have highlighted the potential of phytotherapy in modulating mechanisms related to obesity and depression. Below, selected in vitro and in vivo studies are reviewed, focusing on the bioactivity of plant-derived compounds.

7.1. In Vitro Studies

In vitro studies have consistently demonstrated that specific bioactive compounds inhibit adipogenesis, lipogenesis, and inflammation in adipocytes. For instance, Chen et al. [171] showed that resveratrol (50 µM) significantly reduced 3T3-L1 preadipocyte differentiation by activating AMPK and downregulating adipogenic markers PPARγ and C/EBPα by 45% and 50%, respectively.
Similar anti-adipogenic effects were observed with curcumin, which attenuated lipid accumulation through NF-κB pathway inhibition and the suppression of pro-inflammatory cytokines [172]. Studies using turmeric extracts confirmed reduced cytokine production in macrophages, supporting its anti-inflammatory potential in obesity-related inflammation [173].
EGCG also inhibited 3T3-L1 adipogenesis by downregulating key transcription factors such as PPARγ and C/EBPα and suppressing lipogenic gene expression [174].
Collectively, these findings support the potential of polyphenolic compounds to reduce adipocyte differentiation and inflammatory responses in vitro, justifying their further evaluation in vivo and in clinical trials.
These data also inform the rational design of multi-compound strategies. The high-throughput screening of polyphenols may facilitate the discovery of synergistic combinations. Ultimately, such insights can guide the development of functional food products—e.g., teas, supplements, or bars—that contain bioavailable formulations of curcumin, EGCG, and quercetin.

7.2. In Vivo Studies

In vivo studies play a key role in validating the efficacy of diets enriched with bioactive compounds for managing obesity and depressive symptoms. Several preclinical models have demonstrated dual benefits of natural substances through metabolic and neuroprotective mechanisms.
In a study by Kim et al. [175], turmeric extract (100 mg/kg, 8 weeks) reduced body weight by 15% and serum TNF-α by 35% in HFD-fed rats, with behavioral tests showing improved mood. Similarly, Zhang et al. [176] reported that acai berry extract (200 mg/kg, 6 weeks) decreased body weight (−12%) and cortisol (−30%) while increasing brain serotonin (+25%) in stressed, high-carbohydrate-fed mice.
Khan et al. [177] found that EGCG (50 mg/kg) reduced body weight and visceral fat while improving serotonin and dopamine levels and alleviating depression-like behavior in HFD-fed mice. Omega-3 fatty acids (2 g/kg EPA + DHA) also improved neurochemical and inflammatory profiles—elevating serotonin and reducing IL-6 and TNF-α—alongside benefits in glucose metabolism and insulin sensitivity [178].
Nigella sativa oil (100 mg/kg, 10 weeks) decreased IL-6, reduced body weight by 18%, and improved mood scores in obese rats with depression [179]. Nguyen et al. [180] observed that curcumin (200 mg/kg) lowered cortisol, improved mood-related behavior, and modulated serotonin and dopamine in obese, stressed rats.
These results support translational research toward plant-based, multi-target interventions. For example, EGCG (300 mg/day) combined with a low-glycemic diet could be tested in obese individuals with mild depressive symptoms over 12–16 weeks, assessing outcomes such as body fat percentage, HDRS scores, insulin sensitivity, and BDNF levels.
In summary, natural compounds, such as curcumin, EGCG, anthocyanins, omega-3 fatty acids, and Nigella sativa oil, show synergistic anti-inflammatory, neuroprotective, and mood-regulating effects in vivo, supporting their use in integrated obesity and mental health strategies.

8. Personalized Diets Enriched with Bioactive Compounds and Probiotics in the Management of Obesity and Depression

Based on emerging evidence, individualized diets enriched with selected bioactive compounds offer promising adjunctive support for managing obesity and depression. Their synergistic actions modulate inflammation, neurochemical pathways, and metabolic functions. In their review, Lee et al. [181] emphasized the growing relevance of dietary bioactive compounds—including curcumin, epigallocatechin gallate (EGCG), and omega-3 fatty acids—in managing both obesity and depressive symptoms. These compounds exhibit anti-inflammatory, neuroprotective, and antioxidant properties while modulating cognitive functions and mood [181].
Clinical trials evaluating the efficacy of these compounds typically employed intervention periods ranging from 8 to 12 weeks. These involving curcumin (500–2000 mg/day), EGCG (200–400 mg/day), and omega-3 fatty acids (1–3 g/day) have reported modest weight reduction (1.5–3 kg), lower inflammatory markers (CRP, IL-6), and improved mood assessed by HDRS [181]. Mood improvement was commonly assessed using standardized tools such as the Hamilton Depression Rating Scale (HDRS). The synergistic action of these compounds is thought to result from their combined modulation of inflammation, neurogenesis, and neurotransmitter signaling. Nevertheless, larger and longer-duration clinical trials are warranted to determine optimal dosing strategies and confirm sustained efficacy [181]. Future directions should include the formulation of tailored nutraceuticals combining selected compounds (e.g., EGCG, curcumin, and anthocyanins) with proven safety profiles and bioavailability. These products may serve as adjunct therapies in lifestyle interventions or medical nutrition therapy (MNT) protocols for patients with obesity and comorbid depression.
The development of precision functional food products—such as anti-inflammatory meal kits or ready-to-drink probiotic beverages—could incorporate these compounds. Formulations targeting specific phenotypes (e.g., high BMI with elevated CRP and depressive symptoms) should be explored in personalized nutrition trials. Real-world applications might include lifestyle intervention programs that incorporate phytochemical-rich diets with app-based monitoring.
Preclinical studies confirm the efficacy of curcumin (200 mg/kg), resveratrol (50–100 mg/kg), and herbal extracts, such as ginseng and ashwagandha, in reducing fat mass, enhancing insulin sensitivity, and improving mood-related behaviors in rodent models [182,183].
The role of the gut microbiota in the pathogenesis of both obesity and depression is increasingly recognized. EGCG has been shown to favorably modulate gut microbiota composition, enhance SCFA production, and improve behavioral and metabolic parameters in HFD-fed rodents [184]. The intervention also enhanced short-chain fatty acid (SCFA) production and improved gut barrier integrity, thereby reducing systemic inflammation, a critical mechanism implicated in both disorders [184].
Recent studies highlight the synergistic benefits of combining bioactive compounds with probiotics. The co-administration of curcumin with Lactobacillus rhamnosus and Bifidobacterium longum enhanced weight loss and mood improvement in obese mice [185]. Similarly, a clinical trial combining ginger extract with probiotics led to reduced cortisol levels and improved psychological well-being in obese patients [186].

9. Gut Microbiota Modulation by Bioactive Compounds and Probiotics

In vitro fermentation models have shown that flavonoids from berries (e.g., anthocyanins from blueberries) and phenolic compounds from green tea can modulate gut microbiota by promoting beneficial bacteria, such as Bifidobacterium and Lactobacillus spp., while inhibiting pathogenic species [187,188]. These extracts also increased the production of short-chain fatty acids (SCFAs), particularly butyrate and acetate, which support gut barrier integrity and contribute to gut–brain axis signaling [188].
In vivo studies using rat models of diet-induced obesity revealed that turmeric and green tea extracts increased the abundance of SCFA-producing bacteria such as Faecalibacterium prausnitzii, while reducing levels of obesity-associated Proteobacteria. These microbiota shifts correlated with improved metabolic outcomes, including reduced fasting glucose, better insulin sensitivity, and improved lipid profiles [189].
SCFAs are key mediators of the gut–brain axis that affect neurobehavioral function and mood regulation [190]. Supplementation with plant extracts that enhance SCFA production has been shown to lower cortisol levels and reduce depressive-like behaviors in obese animal models [191]. For example, turmeric extract increased intestinal butyrate and acetate levels in obese rats, which coincided with improvements in behavioral tests related to mood [192].
Evidence also supports the synergistic effects of plant extracts and probiotics. In vivo studies in mice demonstrated that combining Lactobacillus rhamnosus with ginger extract resulted in greater reductions in body weight and improved metabolic markers compared to either treatment alone [193]. This combination enhanced the growth of SCFA-producing bacteria and improved gut barrier function by reducing intestinal permeability and inflammation, which are factors commonly associated with both obesity and depression [194].

10. The Gut–Brain Axis and Translational Potential Perspectives

The interaction among bioactive plant compounds, probiotics, and microbial metabolites plays a crucial role in the gut–brain axis, which is a key regulator of metabolic and neurobehavioral functions. Studies have shown that the modulation of gut microbiota and microbial products, such as SCFAs and tryptophan-derived indoles, can influence neurotransmission, neuroinflammation, and HPA axis function [195].
In a study by Li et al. [196], a combined intervention using probiotics (Lactobacillus rhamnosus and Bifidobacterium longum, 109 colony-forming units (CFU)/day), prebiotics (fructooligosaccharides (FOS) and galactooligosaccharides (GOS)), and plant extracts (turmeric 200 mg/day and green tea 150 mg/day) improved behavioral and metabolic outcomes in animal models with obesity and depression. The treatment led to a ~25% reduction in cortisol levels, lower LDL and triglycerides, and reduced symptoms of anxiety and depression (assessed by open field and avoidance tests). These effects were attributed to the enhanced regulation of the HPA axis and neuroinflammation.
Similarly, Wang et al. [186] conducted a 12-week randomized controlled trial in 120 overweight and obese participants. The intervention included ginger extract (1 g/day) and probiotics (Lactobacillus acidophilus and Bifidobacterium bifidum, 109 CFU/day). Participants who received the combined treatment showed significant improvements: ~25% cortisol reduction, ~3 kg weight loss, decreased IL-6 and TNF-α levels, and better scores on the Beck Depression Inventory-II and Perceived Stress Scale. Waist circumference and body mass index (BMI) also improved.
These findings support the potential of combining specific plant-based compounds with probiotics and prebiotics to target both the metabolic and psychological pathways involved in obesity and depression. The gut–brain axis emerges as a promising therapeutic target for multi-component interventions (Figure 6) [186].

Synbiotic-Based Strategies and Clinical Implementation

The gut–brain axis is increasingly recognized as a bidirectional communication system involving neural, immune, endocrine, and metabolic pathways. This axis plays a significant role in both the pathophysiology of obesity and depression. Alterations in gut microbiota composition—dysbiosis—have been linked to increased gut permeability, systemic inflammation, altered neurotransmitter production, and impaired HPA axis function [179,180].
Preclinical and human studies have shown that specific plant-derived polyphenols and flavonoids can modulate the composition and activity of the gut microbiota, favoring the growth of beneficial bacteria (e.g., Bifidobacterium, Lactobacillus) and suppressing pro-inflammatory taxa [181,182,183]. This modulatory effect is associated with the increased production of short-chain fatty acids (SCFAs), enhanced intestinal barrier integrity, and decreased circulating endotoxins (e.g., LPS).
Importantly, a promising translational strategy involves the combination of targeted probiotics with selected plant polyphenols in the form of synbiotic formulations. Such interventions could exert synergistic effects by modulating both local gut inflammation and systemic immune–neuroendocrine pathways. For instance, anthocyanin-rich extracts co-administered with Lactobacillus rhamnosus strains have been shown to reduce depressive-like behaviors and normalize corticosterone levels in animal models of stress-induced obesity.
From a clinical perspective, these insights could inform the development of next-generation personalized nutrition therapies, integrating gut microbiota profiling with tailored supplementation (e.g., polyphenols + probiotics) in individuals with comorbid depression and metabolic dysfunction. Pilot studies could include outcomes such as microbiota composition (16S rRNA), SCFA levels, CRP, IL-6, and validated mood scales (e.g., PHQ-9, BDI-II).
This microbiota-centered therapeutic approach aligns with the principles of precision medicine and supports the design of non-pharmacological, diet-based adjuncts to traditional care for mood and metabolic disorders.

11. Functional Diets as a Therapeutic Strategy

Recent clinical and preclinical evidence supports the potential of personalized diets enriched with plant-derived bioactive compounds and probiotics in managing obesity accompanied by depressive symptoms. Combinations of curcumin, EGCG, and omega-3 fatty acids have been shown to improve metabolic profiles, mood, and neuroendocrine regulation [181,182]. Similarly, supplementation strategies targeting the gut microbiota contribute to anti-inflammatory effects and the modulation of the hypothalamic–pituitary–adrenal (HPA) axis, with measurable benefits in both metabolic and psychological domains [186,196].
These findings highlight the potential of functional diets or nutraceuticals based on the synergistic actions of bioactive compounds and probiotics. Such interventions may downregulate inflammation, support neurotransmitter balance, regulate HPA activity, and improve energy metabolism—mechanisms relevant to both obesity and depression. While translational clinical trials remain limited, the emerging data point to a promising adjunctive strategy with fewer adverse effects compared to pharmacological monotherapies.
In summary, integrating phytochemicals, probiotics, and microbiota-targeted approaches may offer an effective and well-tolerated therapeutic pathway for patients with obesity and comorbid depression. Continued research into standardized formulations, optimal dosing, and long-term outcomes will be essential for advancing this functional nutrition strategy into clinical practice.
To accelerate translation into practice, public–private collaborations between academic centers, nutraceutical companies, and health systems could initiate pilot programs evaluating the impact of polyphenol- and probiotic-enriched diets. Digital health tools could monitor compliance, mood, and weight changes in real time, integrating these strategies into holistic obesity–depression management frameworks.

Toward Clinical Applications and Functional Product Development

Although multiple in vitro and in vivo studies support the efficacy of bioactive plant compounds in mitigating obesity and depression, translation into clinical practice remains limited. To bridge this gap, structured clinical trials using standardized formulations, validated endpoints (e.g., anthropometric changes, inflammatory markers, and validated psychiatric scales), and well-defined patient groups (e.g., adults with BMI > 30 and elevated PHQ-9 scores) are urgently needed [160,161,162,163,164,165,166,167,168,169,170].
Based on the available evidence, several compounds reviewed in this manuscript—such as epigallocatechin gallate (EGCG), curcumin, resveratrol, genistein, and anthocyanins—are strong candidates for clinical testing as part of functional food interventions or nutraceutical formulations. Bioavailable doses (e.g., EGCG 200–400 mg/day; curcumin 500–1500 mg/day; and anthocyanin-rich extracts 300–500 mg/day) may be incorporated into comprehensive dietary strategies targeting comorbid metabolic and mood disorders.
Potential clinical trial pathways include:
  • 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.
From a functional food development perspective, the creation of microbiota-modulating, polyphenol-enriched products (e.g., flavonoid-fortified snack bars, anthocyanin-based drinks with probiotics) represents a promising translational route. These innovations could be informed by nutrigenomics, metabolomic screening, and gut microbiota profiling, supporting personalized, anti-inflammatory, and mood-regulating dietary protocols.
Furthermore, growing patient interest in dietary interventions reinforces the need to develop evidence-based nutritional guidelines for clinical use. Future research should also prioritize long-term safety, efficacy in diverse populations, bioavailability enhancement strategies (e.g., nanoencapsulation and emulsifiers), and regulatory compliance with EFSA and FDA standards [180,181,182,183,184].

12. Pharmacokinetics and the Potential Side Effects of Selected Phyto-Chemicals: Potential Adverse Effects of Major Compounds (Curcumin, EGCG, and Resveratrol)

In the context of the expanded use of phytochemicals, such as curcumin, EGCG and resveratrol, it is important to understand their pharmacokinetics and the potential risks associated with their use. The following is a detailed review of available data based on the scientific literature.

12.1. Adverse Effects of Resveratrol

Resveratrol (RE) is a widely studied polyphenol with antioxidant, anti-inflammatory, cardioprotective, and neuroprotective properties. Despite promising preclinical outcomes, clinical evidence on its effectiveness remains mixed, and studies on its long-term safety are limited [197].
One of the main challenges in using RE therapeutically is its poor oral bioavailability, which is largely due to its low water solubility (<0.05 mg/mL), rapid metabolism, and extensive conjugation. Although about 70% of ingested RE is absorbed, plasma levels of the unmetabolized compound remain extremely low (<5.0 ng/mL after a 25 mg dose) [197]. Zupančič et al. [198] reported pH-dependent solubility, with optimal values around a pH level of 1.2 and reduced solubility above a pH level of 7.4. RE also shows limited stability at neutral or alkaline pH and degrades under light and oxygen exposure [198,199,200,201].
In vivo, RE binds to albumin, LDL, and integrins, potentially enhancing its tissue distribution and bioactivity [202]. Its metabolites—glucuronides and sulfates—circulate longer than the parent compound and may act as reservoirs for sustained release [203,204].
Toxicological concerns primarily arise at high doses. RE may interfere with cytochrome P450 enzymes, leading to drug interactions and endocrine disruption. High concentrations have been associated with thyroid nodules, pro-oxidant activity, and apoptosis in normal cells via reactive metabolites like o-quinones [205,206,207].
Clinical reports include side effects such as gastrointestinal distress (nausea, diarrhea, and rectal pruritus), elevated liver enzymes (e.g., ALT), leukopenia, and hypersensitivity reactions at doses between 2 and 5 g/day [208,209,210]. Patients with thyroid disorders may be at increased risk of adverse outcomes.
Animal studies confirm dose-dependent toxicity. Doses ≥50 mg/kg in rodents caused nephropathy, inflammation, and mortality, while doses ≤25 mg/kg appeared safe [211,212]. In zebrafish embryos, high doses of RE led to developmental abnormalities and cardiovascular defects [213,214].
In conclusion, while resveratrol is considered safe at moderate levels, high doses or long-term use may lead to adverse effects, including oxidative stress, organ damage, and hormonal dysregulation. Further clinical and toxicological studies are essential to define safe therapeutic ranges and understand their systemic impact [203,204,215,216,217,218].

12.2. Adverse Effects of Epigallocatechin Gallate

Adverse effects of epigallocatechin gallate (EGCG) include a range of toxic effects that can occur with excessive intake or as a result of long-term use of the compound. The literature emphasizes that high doses of EGCG can cause serious liver damage, which has been confirmed in both animal studies and clinical reports. Clinical signs associated with hepatotoxicity include elevated levels of liver enzymes such as transaminases (ALT and AST), as well as the presence of jaundice, abdominal pain, and increased risk of jaundice [217,218,219]. Evidence suggests that the liver is a major target organ for the toxic effects of EGCG, mainly due to its role in metabolizing the compound, as evidenced by the high concentrations of EGCG detected in liver tissue after oral administration [220]. At the cellular level, EGCG exhibits cytotoxic effects on hepatocytes, causing damage to cell membranes and mitochondrial potentials, resulting in reduced cell function and cell death [221,222]
Experimental studies have shown that high doses of EGCG can induce oxidative stress, leading to lipid peroxidation, as evidenced by increases in markers such as MDA and 4-HNE in liver samples [218]. These mechanisms involve activation of the Nrf2-ARE pathway, which under conditions of EGCG excess may act as a defensive response, but at the same time indicates overloading of the body’s an-oxidative systems [223].In addition, excess EGCG can lead to the two-boosting of thiol conjugates, such as EGCG-2′-cysteinyl, which are biomarkers of oxidative stress and chemical damage caused by pro-oxidant compounds [224,225].
The pharmacokinetics of EGCG are characterized by low bioavailability, ranging from 0.2% to 2%, meaning that most of the ingested compound is degraded in the gut by the microbiota or metabolized in the liver [226,227].This indicates that even at high doses, which may exceed safe levels, a small amount of active EGCG can accumulate in the body, increasing the risk of side effects. The safety of EGCG use is therefore related to the dose, the time of administration and the individual’s ability to metabolize the compound, and exceeding established tolerance levels can result in serious health effects, mainly related to liver and kidney function [228,229].
In addition to its effects on the liver and kidneys, EGCG also shows adverse effects on other organs and tissues, mainly due to its pro-oxidant properties. For example, animal studies conducted by Chu et.al. [230] showed that EGCG can harm eye tissues because it causes oxidative stress in various parts of the eye, such as the plasma, aqueous fluid, vitreous body, cornea and retina. High doses of EGCG increase the level of the enzyme SOD1, while suppressing the expression of catalase (CAT), which leads to an excess of reactive oxygen species and oxidative stress in the retina. As a result, the neutralization of ROS to H2O2 is faster, but the decrease in CAT activity hinders the removal of excess H2O2, exacerbating oxidative stress [230].
Oxidative stress can damage pancreatic islet β cells and reduce tissue insulin sensitivity, which may contribute to the development of type 1 diabetes [231]. In rats with streptozotocin (STZ)-induced diabetes, four-day administration of EGCG at 5 mg/kg/intramuscularly aggravated β-cell damage and reduced their response to high glucose concentrations. In this context, vitamin E, which has antioxidant activity, blocked these deleterious effects, suggesting that EGCG acted as a pro-oxidant [232].
In addition, in vitro studies have shown that EGCG is toxic to embryos. At concentrations ranging from 25 to 50 μM, EGCG markedly increased apoptosis in mouse blastocysts, reduced cell number, and interfered with subsequent embryonic development [83]. This damage was related to intrinsic apoptotic signaling processes, as specific inhibitors of caspase-9 and caspase-3 effectively blocked these effects, indicating a pro-oxidant mechanism of EGCG leading to apoptosis.
In terms of effects on DNA, EGCG has shown the ability to damage genetic material. In healthy human lymphocytes, concentrations of 1 to 100 μM caused DNA strand breaks [233], and other studies have confirmed that in a similar dose range (10–100 μM), EGCG causes DNA damage, which is related to the substance’s prooxidant effects [84]. It is worth mentioning that despite these actions, EGCG does not show clastogenic effects in in vivo studies [234].
In addition, in fat cells, EGCG induces oxidative stress, which can have negative health effects by interfering with hormone function in adipocytes [235]. In conclusion, although EGCG is mainly known as an antioxidant, its pro-oxidant effects can lead to cellular, DNA, and tissue damage, which is important in the context of the potential adverse effects of this substance. Excessive consumption of epigallocatechin gallate is associated with the risk of hepatotoxicity, nephrotoxicity, and other adverse effects, which calls for caution and monitoring of doses in the context of use as a dietary supplement.

12.3. Adverse Effects of Curcumin

Although curcumin is widely considered safe and is widely used as a dietary ingredient and supplement, there is evidence of its potential toxicity at certain conditions and doses. In vitro and in vivo studies have shown that curcumin can induce DNA damage and chromosomal aberrations at concentrations similar to those that show beneficial effects (2.5–5 µg/mL). Such effects, including induction of mutations and genetic damage, have been linked to carcinogenesis, raising concerns about the safety of long-term use [230,236,237,238,239,240,241].
Long-term studies in animal models have shown that intake of curcumin in high doses can lead to pathological changes such as inflammation, hyperplasia and ulceration in the gastrointestinal tract, as well as hyperplasia of thyroid cells and increased risk of cancer in some organs [242]. Notably, consumption of diets containing turmeric extract (containing 79–85% curcumin) over a period of up to 2 years increased the incidence of tumors, suggesting a potential mutagenic or tumor-promoting effect [242,243].
Curcumin can generate reactive oxygen species (ROS) in high concentrations, which contributes to oxidative stress and DNA damage. In addition, the presence of a 2,4-en-xetone group in its chemical structure enables Michael addition-type reactions that can modify key proteins and enzymes, including topoisomerase II, p53 and antioxidant enzymes such as thioredoxin-reductase, which can lead to uncontrolled cancer cell growth [244,245,246,247,248].
Curcumin shows poor bioavailability after oral administration due to its intensive metabolism in the intestines and liver. Studies have shown that after large doses (e.g., 10–12 g) in studies on healthy volunteers, only one person could detect the presence of free curcumin in plasma at nanomolar levels, which is well below the concentrations needed to induce cytotoxic effects [249,250]. In addition, curcumin is metabolized mainly to glucuronides and sulfates, which limits its availability in some tissues and organs [249,251,252].
Moreover, curcumin has the ability to inhibit drug-metabolizing enzymes such as cytochrome P450, glutathione-S-transferase and UDP-glucuronosyltransferase. This can lead to increased concentrations of certain drugs in the body, which increases the risk of side effects [253,254,255,256,257,258].
According to reports from international organizations such as JECFA and EFSA, the safe daily dose of curcumin is up to 3 mg per kilogram of body weight. In studies on healthy individuals using doses of 0.45 to 3.6 grams per day for up to four months, mainly mild side effects such as nausea, diarrhea, headaches and increased liver enzyme levels were reported [259,260]. At higher doses, especially above 8 g per day, more serious effects may occur, although the data are limited and long-term safety is not yet fully confirmed [260].
In conclusion, despite the widespread belief in curcumin’s safety as a dietary supplement, there is growing evidence of its potential harm, especially at high doses and long-term use. Cases of DNA-damaging effects, tumor promotion and serious pathological changes in animal models indicate that its use must be cautious and preceded by thorough safety studies. In addition, its low bioavailability limits therapeutic efficacy, necessitating strategies that increase absorption, but which may also increase the risk of side effects.
Summarizing the most significant adverse effects on the body, it should be pointed out that curcumin can induce DNA damaging and chromosomal aberrations at the in vitro and in vivo levels, which may have oncogenic implications [236,237,238,239,240,241].
Long-term animal studies suggest that high doses can lead to pathological changes and increase the risk of cancer [242,243]. Mechanisms of deleterious effects include ROS generation and Michael addition-type reactions that can modify key proteins and enzymes [244,246,247,248].
In addition, low bioavailability after oral administration limits the achievement of effective tissue concentrations [249]. Curcumin can inhibit drug-metabolizing enzymes, which increases the risk of pharmacological interactions [253,254,255,256].
In addition, long-term use of high doses (e.g., more than 8 g per day) can lead to side effects such as nausea, diarrhea, increased liver enzymes, and iron deficiency [260,261].

13. Limitations

This scoping review has several limitations. First, the review was not registered in a systematic review registry, which may limit transparency compared to pre-registered protocols. Second, although the PRISMA guidelines were followed, no formal assessment of risk of bias was conducted for the included studies, as the focus was on mechanistic diversity and exploratory synthesis rather than quality-weighted meta-analysis. Third, only studies published in English were included, which may introduce language bias. Additionally, the inclusion of heterogeneous study types (in vitro, in vivo, and clinical) without stratification may affect the comparability of findings. Lastly, publication bias could not be excluded, as studies with negative results may have been underrepresented.

14. Conclusions

The comorbidity of obesity and depression represents a major clinical and public health challenge, which is characterized by overlapping mechanisms such as chronic inflammation, oxidative stress, neurotransmitter imbalances, and gut–brain axis disruption. This review underscores the therapeutic potential of plant-derived bioactive compounds—particularly polyphenols, flavonoids, alkaloids, and betalains—in targeting both metabolic dysfunction and mood disorders.
The key mechanisms of action include the reduction of neuroinflammation, the modulation of serotonergic and dopaminergic pathways, the enhancement of neurotrophic factors (e.g., BDNF), the suppression of adipogenesis, and the improvement of insulin sensitivity. Compounds such as quercetin, genistein, EGCG, curcumin, and anthocyanins have shown promising results in preclinical models and emerging evidence from human studies.
Incorporating these phytochemicals into functional diets or adjunct therapies may offer a safe, integrative strategy for patients with coexisting obesity and depressive symptoms. However, to facilitate clinical translation, future research should prioritize standardized dosing, long-term safety assessments, and synergistic combinations, especially those involving microbiota-targeted interventions.
Ultimately, integrating phytotherapy with personalized nutrition and microbiota modulation may redefine future therapeutic approaches for obesity-related mood disorders.

Author Contributions

Conceptualization, S.L.-W.; methodology, K.S.; software, K.S.; validation, K.S. and S.L.-W.; formal analysis, K.S.; investigation, K.S.; resources, K.S.; data curation, K.S.; writing—original draft preparation, K.S.; writing—review and editing, S.L.-W.; visualization, K.S. and S.L.-W.; supervision, S.L.-W.; project administration, S.L.-W.; funding acquisition, S.L.-W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Journal Molecules, MDPI.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Chan, M. Obesity and Diabetes: The Slow-Motion Disaster. Milbank Q. 2017, 95, 11–14. [Google Scholar] [CrossRef] [PubMed]
  2. Schwarzenberg, S.J.; Sinaiko, A.R. Obesity and inflammation in children. Paediatr. Respir. Rev. 2006, 7, 239–246. [Google Scholar] [CrossRef]
  3. 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]
  4. 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]
  5. 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]
  6. Shelton, R.C.; Miller, A.H. Inflammation in depression: Is adiposity a cause? Dialogues Clin. Neurosci. 2011, 13, 41–53. [Google Scholar] [CrossRef]
  7. 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]
  8. 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]
  9. Olszanecka-Glinianowicz, M. Depression—Cause or result of obesity? Endokrynol. Otyłość Zab. Przem. Mat. 2008, 4, 78–85. [Google Scholar]
  10. 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]
  11. Kazimierska, I. Otyłość to choroba, a nie defekt estetyczny. Kurier Medyczny 2021, 2, 14–15. [Google Scholar]
  12. Kopacz, M.S.; Wichowicz, H.M. Obesity—Causes, Types, Treatment, Effects. Eng. Sci. Technol. 2018, 4, 65–79. [Google Scholar] [CrossRef]
  13. Stunkard, A.J.; Faith, M.S.; Allison, K.C. Depression and obesity. Biol. Psychiatry 2003, 54, 330–337. [Google Scholar] [CrossRef] [PubMed]
  14. Mayorga, P. El Adipocito Como Órgano Endocrino: Implicaciones Fisiopatológicas y Terapéuticas. Rev. Med. 2007, 152, 225–242. [Google Scholar]
  15. Stolarczyk, E. Adipose tissue inflammation in obesity: A metabolic or immune response? Curr. Opin. Pharmacol. 2017, 37, 35–40. [Google Scholar] [CrossRef]
  16. 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]
  17. Stalikas, C.D. Extraction, separation, and detection methods for phenolic acids and flavonoids. J. Sep. Sci. 2007, 30, 3268–3295. [Google Scholar] [CrossRef]
  18. 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]
  19. 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]
  20. 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]
  21. 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]
  22. 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]
  23. 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]
  24. 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]
  25. 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]
  26. 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]
  27. 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]
  28. 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]
  29. 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]
  30. 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]
  31. 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]
  32. 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]
  33. 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]
  34. 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]
  35. 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]
  36. 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]
  37. 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]
  38. 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]
  39. 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]
  40. 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]
  41. 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]
  42. 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]
  43. 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]
  44. 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]
  45. 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]
  46. 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]
  47. Lee, G.; Bae, H. Therapeutic Effects of Phytochemicals and Medicinal Herbs on Depression. BioMed Res. Int. 2017, 2017, 6596241. [Google Scholar] [CrossRef]
  48. Nurzyńska-Wierdak, R. Plants with Potential Importance in Supporting the Treatment of Depression: Current Trends, and Research. Pharmaceuticals 2024, 17, 1489. [Google Scholar] [CrossRef]
  49. Kalueff, A.V.; Nutt, D.J. Role of GABA in anxiety and depression. Depress. Anxiety 2007, 24, 495–517. [Google Scholar] [CrossRef] [PubMed]
  50. 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]
  51. Kussmann, M.; Abe Cunha, D.H.; Berciano, S. Bioactive compounds for human and planetary health. Front. Nutr. 2023, 10, 1193848. [Google Scholar] [CrossRef]
  52. 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]
  53. Murawska-Ciałowicz, E. Adipose tissue—Morphological and biochemical characteristic of different depots. Postepy Hig. Med. Dosw. 2017, 71, 466–484. [Google Scholar] [CrossRef] [PubMed]
  54. Kolb, H. Obese visceral fat tissue inflammation: From protective to detrimental? BMC Med. 2022, 20, 494. [Google Scholar] [CrossRef]
  55. 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]
  56. 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]
  57. 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]
  58. 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]
  59. Williamson, G. The Role of Polyphenols in Modern Nutrition. Nutr. Bull. 2017, 42, 226–235. [Google Scholar] [CrossRef]
  60. 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]
  61. Bravo, L. Polyphenols: Chemistry, Dietary Sources, Metabolism, and Nutritional Significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef] [PubMed]
  62. 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]
  63. 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]
  64. 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]
  65. 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]
  66. 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]
  67. Singh, M.; Thrimawithana, T.; Shukla, R.; Adhikari, B. Managing obesity through natural polyphenols: A review. Future Foods 2020, 1–2, 100002. [Google Scholar] [CrossRef]
  68. 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]
  69. 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]
  70. 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]
  71. 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]
  72. 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]
  73. 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]
  74. 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]
  75. 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]
  76. 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]
  77. 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]
  78. 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]
  79. 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]
  80. 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]
  81. 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]
  82. 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]
  83. 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]
  84. 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]
  85. 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]
  86. 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]
  87. 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]
  88. 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]
  89. 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]
  90. 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]
  91. 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]
  92. 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]
  93. 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]
  94. 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]
  95. 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]
  96. 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]
  97. 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]
  98. Albani, D.; Polito, L.; Signorini, A.; Forloni, G. Neuroprotective properties of resveratrol in different neurodegenerative disorders. Biofactors 2010, 36, 370–376. [Google Scholar] [CrossRef]
  99. Gu, Z.; Chu, L.; Han, Y. Therapeutic effect of resveratrol on mice with depression. Exp. Ther. Med. 2019, 18, 3015–3020. [Google Scholar] [CrossRef]
  100. 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]
  101. 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]
  102. 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]
  103. 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]
  104. 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]
  105. 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]
  106. 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]
  107. 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]
  108. 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]
  109. 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]
  110. 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]
  111. Habtemariam, S.; Varghese, G.K. The antidiabetic therapeutic potential of dietary polyphenols. Curr. Pharm. Biotechnol. 2014, 15, 391–400. [Google Scholar] [CrossRef] [PubMed]
  112. 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]
  113. 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]
  114. 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]
  115. 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]
  116. 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]
  117. 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]
  118. Wybraniec, S.; Mizrahi, Y. Fruit flesh betacyanin pigments in Hylocereus cacti. J. Agric. Food Chem. 2002, 50, 6086–6089. [Google Scholar] [CrossRef] [PubMed]
  119. 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]
  120. Nakamura, K.; Fuster, J.J.; Walsh, K. Adipokines: A link between obesity and cardiovascular disease. J. Cardiol. 2014, 63, 250–259. [Google Scholar] [CrossRef]
  121. 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]
  122. 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]
  123. 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]
  124. 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]
  125. 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]
  126. 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]
  127. 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]
  128. 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]
  129. 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]
  130. Szafrański, T. Herbal remedies in depression—State of the art. Psychiatr. Pol. 2014, 48, 59–73. [Google Scholar] [CrossRef]
  131. 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]
  132. 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]
  133. 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]
  134. 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]
  135. 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]
  136. Rabiei, Z.; Rabiei, S. A review on antidepressant effect of medicinal plants. Bangladesh J. Pharmacol. 2017, 12, 1–11. [Google Scholar] [CrossRef]
  137. 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]
  138. 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]
  139. 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]
  140. 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]
  141. 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]
  142. 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]
  143. 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]
  144. 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]
  145. 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]
  146. 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]
  147. 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]
  148. 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]
  149. 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]
  150. 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]
  151. Andrade, C. A Critical Examination of Studies on Curcumin for Depression. J. Clin. Psychiatry 2014, 75, e1110–e1112. [Google Scholar] [CrossRef] [PubMed]
  152. 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]
  153. 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]
  154. 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]
  155. 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]
  156. 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]
  157. 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]
  158. 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]
  159. 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]
  160. 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]
  161. 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]
  162. 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]
  163. 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]
  164. 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]
  165. 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]
  166. 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]
  167. 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]
  168. 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]
  169. 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]
  170. 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]
  171. 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]
  172. 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]
  173. 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]
  174. 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]
  175. 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]
  176. 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]
  177. 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]
  178. 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]
  179. 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]
  180. 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]
  181. Lee, B.; Kim, C.Y. Dietary bioactive compounds for managing obesity and depression: A clinical perspective. Nutrients 2021, 13, 1254. [Google Scholar]
  182. 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]
  183. 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]
  184. 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]
  185. 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]
  186. 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]
  187. 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]
  188. 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]
  189. 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]
  190. 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]
  191. 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]
  192. 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]
  193. 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]
  194. 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]
  195. 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]
  196. 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]
  197. 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]
  198. 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]
  199. 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]
  200. 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]
  201. 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]
  202. 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]
  203. 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]
  204. 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]
  205. 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]
  206. 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]
  207. 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]
  208. 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]
  209. 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]
  210. Muñoz, O.; Bustamante, S. Pharmacological Properties of Resveratrol. A Pre-Clinical and Clinical Review. Biochem. Pharmacol. Open Access 2015, 4, 1000184. [Google Scholar] [CrossRef]
  211. 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]
  212. 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]
  213. 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]
  214. 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]
  215. 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]
  216. 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]
  217. 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]
  218. 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]
  219. 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]
  220. 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]
  221. Tachibana, H. Green tea polyphenol sensing. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2011, 87, 66–80. [Google Scholar] [CrossRef] [PubMed]
  222. 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]
  223. 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]
  224. 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]
  225. 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]
  226. 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]
  227. 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]
  228. 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]
  229. 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]
  230. 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]
  231. 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]
  232. 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]
  233. Khan, N.; Mukhtar, H. Tea and health: Studies in humans. Curr. Pharm. Des. 2013, 19, 6141–6147. [Google Scholar] [CrossRef]
  234. da Silva Pinto, M. Tea: A new perspective on health benefits. Food Res. Int. 2013, 53, 558–567. [Google Scholar] [CrossRef]
  235. 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]
  236. 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]
  237. 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]
  238. 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]
  239. 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]
  240. 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]
  241. 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]
  242. 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]
  243. 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]
  244. 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]
  245. 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]
  246. 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]
  247. 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]
  248. 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]
  249. 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]
  250. 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]
  251. 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]
  252. 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]
  253. Mancuso, C.; Barone, E. Curcumin in clinical practice: Myth or reality? Trends Pharmacol. Sci. 2009, 30, 333–334. [Google Scholar] [CrossRef]
  254. 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]
  255. 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]
  256. 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]
  257. 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]
  258. Means, R.T., Jr. Ironing out complementary medicine. Blood 2009, 113, 270–271. [Google Scholar] [CrossRef] [PubMed]
  259. 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]
  260. 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]
  261. Gubin, M.M.; Vesely, M.D. Cancer Immunoediting in the Era of Immuno-oncology. Clin. Cancer Res. 2020, 28, 3917–3928. [Google Scholar]
Figure 1. PRISMA-ScR flow diagram illustrating the study selection process. After the removal of duplicates and the application of inclusion and exclusion criteria, 261 publications were included in the final analysis.
Figure 1. PRISMA-ScR flow diagram illustrating the study selection process. After the removal of duplicates and the application of inclusion and exclusion criteria, 261 publications were included in the final analysis.
Molecules 30 02827 g001
Figure 2. Bidirectional pathophysiological links between obesity and depression mediated by inflammation, oxidative stress, and gut dysbiosis. Polyphenolic compounds counteract these shared mechanisms through anti-inflammatory, antioxidant, microbiota-modulating, neuroprotective, and metabolic effects.
Figure 2. Bidirectional pathophysiological links between obesity and depression mediated by inflammation, oxidative stress, and gut dysbiosis. Polyphenolic compounds counteract these shared mechanisms through anti-inflammatory, antioxidant, microbiota-modulating, neuroprotective, and metabolic effects.
Molecules 30 02827 g002
Figure 3. Molecular mechanisms through which selected bioactive compounds (e.g., quercetin, genistein, and curcumin) act on the brain–gut–adipose axis to counteract inflammation, enhance AMPK activity, and modulate serotonin and neuroinflammatory pathways.
Figure 3. Molecular mechanisms through which selected bioactive compounds (e.g., quercetin, genistein, and curcumin) act on the brain–gut–adipose axis to counteract inflammation, enhance AMPK activity, and modulate serotonin and neuroinflammatory pathways.
Molecules 30 02827 g003
Figure 4. Synergistic actions of plant bioactive compounds on AMPK and BDNF signaling pathways, contributing to adipocyte reduction, improved insulin sensitivity, and neuroprotection.
Figure 4. Synergistic actions of plant bioactive compounds on AMPK and BDNF signaling pathways, contributing to adipocyte reduction, improved insulin sensitivity, and neuroprotection.
Molecules 30 02827 g004
Figure 5. Flavonoid-mediated modulation of neuroinflammation, oxidative stress, and BDNF expression in relation to depressive symptoms.
Figure 5. Flavonoid-mediated modulation of neuroinflammation, oxidative stress, and BDNF expression in relation to depressive symptoms.
Molecules 30 02827 g005
Figure 6. Synergistic interactions among plant bioactives, probiotics, and gut microbiota in modulating inflammation, metabolism, and neurobehavioral outcomes through gut–brain axis mechanisms.
Figure 6. Synergistic interactions among plant bioactives, probiotics, and gut microbiota in modulating inflammation, metabolism, and neurobehavioral outcomes through gut–brain axis mechanisms.
Molecules 30 02827 g006
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.

Share and Cite

MDPI and ACS Style

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

AMA Style

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 Style

Sochacka, 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 Style

Sochacka, 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

Article Metrics

Back to TopTop