Next Article in Journal
Identification of Novel Genetic Variants in a Cohort of Congenital Hypogonadotropic Hypogonadism: Computational Analysis of Pathogenicity Predictions
Previous Article in Journal
The Predictive Impact of HPV Genotypes, Tumor Suppressors and Local Immune Response in the Regression of Cervical Intraepithelial Neoplasia 2-3: A Prospective Population-Based Cohort Study
Previous Article in Special Issue
Cornus mas L. Extracts Exhibit Neuroprotective Properties, Further Enhanced by Metal-Bound Energy-Linked Organic Substrates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications

1
Department of Functional Morphology, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan
2
Department of Community Health Care and Sciences, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose 204-8588, Tokyo, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(11), 5206; https://doi.org/10.3390/ijms26115206
Submission received: 28 February 2025 / Revised: 23 May 2025 / Accepted: 27 May 2025 / Published: 28 May 2025
(This article belongs to the Special Issue Natural Compounds in Human Health and Disease)

Abstract

Chronic inflammation is a key contributor to the development and progression of numerous chronic diseases, including cardiovascular diseases, type 2 diabetes, neurodegenerative disorders, cancer, and obesity. As the side effects of conventional anti-inflammatory drugs pose challenges, plant-derived compounds have emerged as promising alternatives due to their potent anti-inflammatory properties and minimal adverse effects. This review explores the molecular mechanisms by which these compounds alleviate chronic inflammation and highlights their potential role in disease prevention. Polyphenols (e.g., quercetin and resveratrol), flavonoids (e.g., luteolin and apigenin), carotenoids (e.g., β-carotene and lycopene), and other phytochemicals (e.g., curcumin and gingerol) modulate inflammatory pathways, such as nuclear factor-κB and mitogen-activated protein kinase, reduce oxidative stress, and inhibit pro-inflammatory cytokines. Plant-derived compounds interact with the gut microbiota, enhancing anti-inflammatory effects. Evidence from animal studies and clinical trials has demonstrated their efficacy in reducing inflammation-related biomarkers and improving health outcomes. However, challenges such as low bioavailability and determination of the optimal dosage require further investigation. Advancing delivery technologies and personalized nutrition strategies may help overcome these barriers. This review emphasizes the therapeutic potential of plant-derived compounds in preventing chronic diseases and underscores the need for continued research to translate these findings into practical applications for public health.

1. Introduction

Chronic inflammation is considered one of the most important and complex health challenges in modern medicine [1,2]. Chronic inflammation is a low-grade inflammatory condition caused by the body’s exposure to persistent irritation and stress, which, when sustained over time, promotes the progression of many major chronic diseases, including cardiovascular disease (CVD), metabolic syndromes, type 2 diabetes, Alzheimer’s disease, Parkinson’s disease, and cancer [3,4,5]. Behind this chronic inflammation lies a persistent over-activation of the immune response, persistent overproduction of inflammatory cytokines (interleukin-1β [IL-1β], interleukin-6 [IL-6], tumor necrosis factor-α [TNF-α], etc.), overproduction of reactive oxygen species (ROS), and associated cellular damage and tissue dysfunction [6,7,8]. These factors interact to form a vicious cycle of inflammation that accelerates disease onset and progression.
For example, in CVDs, chronic inflammation causes dysfunction of vascular endothelial cells and promotes the progression of atherosclerosis [9,10]. Furthermore, inflammatory cytokines have been shown to disrupt blood pressure regulation and induce vascular wall remodeling [11,12]. On the other hand, in metabolic syndromes, type 2 diabetes, and chronic obesity, chronic adipose tissue and hepatic inflammation exacerbate insulin resistance and disrupt glucose metabolism [13,14,15]. In neurodegenerative diseases, chronic inflammation in the brain leads to neuronal damage and apoptosis and is deeply involved in the progression of Alzheimer’s disease and Parkinson’s disease [16]. In cancer, chronic inflammation has been reported to alter the tumor microenvironment and promote cancer cell growth and metastasis [17,18]. Chronic inflammation triggers these diseases and is a factor in disease progression.
In modern medicine, chemical therapies and pharmaceuticals are used to control chronic inflammation [19]. For example, nonsteroidal anti-inflammatory drugs (NSAIDs) and steroids are widely used to control inflammation because they inhibit the production of inflammatory cytokines [20,21]. However, these therapies are associated with side effects and risks with long-term use; NSAIDs can cause gastrointestinal disturbances and impaired renal function, whereas steroids cause immunosuppression and metabolic abnormalities [22,23]. Therefore, new approaches are needed to control inflammation safely and effectively. Against this background, plant-derived compounds have attracted attention (Figure 1). These compounds are extracted from naturally occurring foods, medicinal herbs, and spices and are expected to have diverse health effects, including anti-inflammatory, antioxidant, immunomodulatory, and even cancer prevention effects. In particular, polyphenols (e.g., quercetin and resveratrol), flavonoids (e.g., luteolin and apigenin), carotenoids (e.g., β-carotene and lycopene), and other plant compounds (e.g., curcumin and gingerol) have proven their anti-inflammatory effects in numerous studies [24,25,26,27,28,29].
The molecular mechanisms by which plant-derived compounds exert their anti-inflammatory effects are diverse [30,31]. These compounds not only suppress the activity of key inflammatory signaling pathways, such as nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK), but also reduce oxidative stress and prevent cell damage by decreasing ROS [32,33]. They have also been shown to disrupt the vicious cycle of inflammation by suppressing the production of inflammatory cytokines [34]. Furthermore, through their interaction with intestinal microflora, it has been suggested that they may reduce systemic inflammation [35]. Therefore, plant-derived compounds are considered promising tools in the prevention and management of chronic diseases.
This study provides a comprehensive review of how plant-derived compounds contribute to the prevention of chronic inflammation and related diseases. Specifically, the molecular mechanisms underlying the anti-inflammatory effects of these compounds, results from disease models and clinical studies, and the challenges and prospects for their practical application are detailed. The potential role of plant-derived compounds in medicine and public health is discussed, and their potential for the next generation of inflammation therapy and disease prevention is explored.

2. Materials and Methods

For this review, literature searches were conducted using the electronic databases PubMed, ScienceDirect, and Web of Science, with searches focusing on recent data. Search terms included the following: “plant-derived compound”, “inflammatory”, “oxidative stress”, “chronic disease”, “neurodegenerative disease”, “cancer”, polyphenol”, “flavonoid”, “carotenoid”, “curcumin”, and “gingerol”. Duplicate references were removed. Based on this search and criteria, the cited articles were refined for this review.

3. Chronic Inflammation and Disease Relationships

3.1. Mechanisms of Chronic Inflammation

Chronic inflammation is caused by repeated or prolonged exposure to internal and external stressors. These factors include infections, persistent trauma, lifestyle habits such as obesity and inactivity, and environmental factors such as smoking and air pollution [36,37,38,39,40]. These factors can lead to an uncontrolled immune response and cause the body to undergo the following characteristic processes.

3.1.1. Overproduction of Inflammatory Cytokines

Inflammatory cytokines (such as IL-1β, IL-6, and TNF-α) are important molecules produced by the immune system to enhance inflammatory responses [41,42,43]. In a normal immune response, these molecules play an important role in repairing the site of infection or injury, but in conditions of chronic inflammation, they are overproduced and attack healthy cells and tissues. As a result, local tissue damage leads to systemic pathological changes.

3.1.2. Oxidative Stress and ROS Generation

During the inflammatory process, immune cells (especially neutrophils and macrophages) produce ROS [44,45]. While ROS attacks pathogens, excessive ROS accumulation damages cell membranes, proteins, and DNA, and DNA damage can lead to cell death and mutations, contributing to an increased risk of cancer [46]. In addition, oxidative stress further activates chronic inflammatory pathways, creating a vicious cycle of inflammation [47].

3.1.3. Activation of Inflammatory Signaling Pathways

In states of chronic inflammation, key intracellular signaling pathways are homeostatically activated. In particular, NF-κB, MAPK, and Janus kinase/signaling and transcription activator pathways are involved [48,49]. These pathways induce the expression of inflammatory cytokines, chemokines, prostaglandins, and other inflammatory molecules [50,51,52]. This process contributes significantly to the persistence of pathologic inflammation.

3.1.4. Immune Cell Hyperactivation and Tissue Infiltration

In chronic inflammatory conditions, immune cells (macrophages, microglia, neutrophils, etc.) are over-activated and persistent infiltration of the inflammatory site is observed [53]. These immune cells produce additional inflammatory mediators that also damage surrounding healthy tissue [54]. As this process progresses, inflammation becomes the primary cause of autoimmune and chronic diseases.

3.1.5. Polyphenol-Mediated Regulation of Antioxidant Enzymes: Focus on Catalase

Polyphenols exert strong antioxidant effects by directly scavenging ROS and modulating the expression and activity of endogenous antioxidant enzymes, such as catalase, superoxide dismutase, and glutathione peroxidase [55,56]. Among these, catalase plays a crucial role in the detoxification of hydrogen peroxide (H2O2), a major ROS involved in oxidative stress and chronic inflammation.
Several studies have demonstrated that polyphenols, including resveratrol, quercetin, epigallocatechin gallate (EGCG), and curcumin, can enhance catalase activity through transcriptional and post-translational mechanisms [57,58]. For example, resveratrol has been shown to upregulate catalase gene expression via the activation of the Nrf2 signaling pathway, a master regulator of antioxidant response [59]. Similarly, quercetin increases catalase activity in hepatic and endothelial cells by promoting Nrf2 nuclear translocation and binding to the antioxidant response element (ARE) in the catalase promoter region [60].
In addition to gene regulation, polyphenols may stabilize catalase protein or protect it from oxidative inactivation. For instance, EGCG and curcumin have been reported to preserve catalase activity under oxidative stress conditions in models of neurodegeneration and metabolic syndrome [60]. These polyphenol-mediated effects on catalase contribute significantly to reductions in cellular ROS levels, maintenance of redox homeostasis, and the prevention of ROS-induced damage in various chronic diseases.

3.2. Role of Inflammation in Chronic Disease

Chronic inflammation is involved in the development and progression of nearly all major chronic diseases, and the mechanisms involved differ for each disease. Chronic inflammation is closely associated with the following: CVDs, metabolic syndromes, type 2 diabetes, neurodegenerative diseases, and chronic obesity. The role of chronic inflammation in these diseases will be discussed in detail.

3.2.1. CVDs

Chronic inflammation is one of the major pathological mechanisms in CVDs, such as atherosclerosis and hypertension [61]. Inflammatory cytokines (especially IL-6 and TNF-α) impair vascular endothelial cell function and increase endothelial cell permeability [62]. In the process, low-density lipoproteins accumulate in the vessel wall and are oxidized, leading to the formation of atherosclerotic plaques [63]. Chronic inflammation also accelerates the progression of atherosclerotic disease by causing a breakdown in blood pressure regulation and promoting vessel wall remodeling [64].

3.2.2. Metabolic Syndromes and Type 2 Diabetes

Chronic inflammation is directly involved in the development and exacerbation of insulin resistance [65]. With obesity, inflammatory cytokines (especially TNF-α and IL-6) secreted by adipose tissue inhibit insulin receptor signaling and cause chronic elevations in blood glucose levels. In addition, adipokine secretion from adipocytes is abnormal in obesity, amplifying chronic systemic inflammation [66]. The result is a progressive disruption of glucose metabolism and an increased risk of diabetic complications. Recent studies have highlighted the role of semaphorin 3E (Sema3E), a member of the semaphorin family originally known for its function in axon guidance, in mediating adipose tissue inflammation and insulin resistance. Sema3E is highly expressed in the adipose tissue of obese mice and humans, particularly by pro-inflammatory macrophages. This protein binds to its receptor PlexinD1, which is also upregulated in inflamed adipose tissues, leading to enhanced macrophage infiltration and reduced egress from adipose depots. This retention of M1-like macrophages sustains local inflammation and increases the production of cytokines such as IL-6 and TNF-α, which further impairs insulin signaling in adipocytes. Moreover, genetic or pharmacological inhibition of Sema3E/PlexinD1 signaling has been shown to attenuate adipose inflammation, improve insulin sensitivity, and normalize glucose tolerance in obese mice, indicating that Sema3E is not only a marker of metabolic inflammation but is also a functional contributor to insulin resistance. These findings suggest that targeting the Sema3E–PlexinD1 axis may provide a novel therapeutic strategy for mitigating inflammation-induced metabolic disorders.

3.2.3. Neurodegenerative Diseases

Brain inflammation is a major factor in the progression of neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease [67]. Microglia, immune cells of the central nervous system (CNS), are chronically activated by pathological stimuli (e.g., amyloid-β accumulation and abnormal alpha-synuclein), releasing inflammatory cytokines and ROS [68]. This inflammatory response leads to neuronal damage and apoptosis and promotes a decline in neurological function [68]. The interaction between brain and vascular inflammation has also been noted to play an important role in the progression of neurodegenerative diseases.

3.2.4. Cancer

Chronic inflammation is a potent promoter of cancer development and progression [69]. Persistent chronic inflammation leads to the secretion of inflammatory cytokines and growth factors (e.g., IL-6, VEGF), which modify the tumor microenvironment [70]. This environment promotes cancer cell proliferation, angiogenesis, and metastasis and protects cancer cells from the immune system. In addition, inflammation-induced oxidative stress and DNA damage can induce mutations and increase carcinogenicity [71,72].
Moreover, the epidermal growth factor receptor (EGFR) signaling pathway plays a critical role in linking chronic inflammation to tumorigenesis. EGFR is frequently overexpressed or mutated in various cancers and is activated by inflammatory mediators such as TNF-α and IL-1β [73]. EGFR activation stimulates downstream signaling cascades, including the RAS-RAF-MEK-ERK and PI3K-AKT pathways, which promote cell proliferation, survival, and resistance to apoptosis [74]. Inflammatory conditions can enhance EGFR signaling directly through ligand induction (e.g., EGF, TGF-α) and indirectly via ROS-mediated receptor activation, further contributing to an environment that is conducive to tumor initiation and progression [75]. Notably, plant-derived compounds such as curcumin, resveratrol, and EGCG have been shown to inhibit EGFR signaling and its downstream pathways, exerting anti-proliferative and anti-inflammatory effects in cancer models [76].

3.2.5. Chronic Obesity

Chronic obesity is characterized by persistent low-grade inflammation, and adipose tissue is known to be the primary starting point of inflammation [77]. As obesity progresses, adipose tissue increases the secretion of inflammatory cytokines and promotes macrophage infiltration, triggering a systemic inflammatory response [78]. This chronic inflammation has been reported to exacerbate insulin resistance and metabolic abnormalities, increasing the risk of type 2 diabetes, CVD, and even cancer and neurodegenerative diseases [79].
Enlarged adipocytes secrete excessive amounts of the inflammatory cytokines (TNF-α, IL-6, and IL-1β) [80,81,82]. These cytokines activate immune cells in adipose tissue and cause them to release more inflammatory mediators, forming a vicious cycle of chronic inflammation. TNF-α, in particular, is known to inhibit insulin signaling and cause abnormal glucose metabolism. IL-6 has been reported to promote the production of C-reactive protein in the liver, which is reflected in the blood as a systemic inflammatory marker [83]. With obesity, adipose tissue becomes infiltrated by macrophages, whose phenotype shifts from anti-inflammatory type M2 to inflammatory type M1 [84]. Type M1 macrophages produce large amounts of inflammatory cytokines, further amplifying adipose tissue inflammation. This accumulation of inflammatory macrophages plays an important role in the pathogenesis of obesity, contributing to adipocyte dysfunction and insulin resistance. Furthermore, macrophage-derived nitric oxide and ROS have been shown to increase adipocyte stress responses and induce apoptosis [85,86]. Chronic inflammation associated with obesity inhibits phosphorylation of insulin receptor substrate-1, an important component of the insulin signaling pathway, thereby reducing insulin action. TNF-α has been shown to decrease insulin receptor substrate-1 activity, particularly via serine/threonine phosphorylation, resulting in impaired glucose uptake and regulation of glucose metabolism [87,88]. This exacerbation of insulin resistance is directly involved in the development of type 2 diabetes and contributes to hyperglycemia and dyslipidemia.
Recent studies suggest that alterations in the gut microbiota are an important factor in obesity-related inflammation [89]. Intestinal permeability is increased in the gut of patients with obesity, facilitating the influx of bacteria-derived endotoxins called lipopolysaccharides into the bloodstream. Lipopolysaccharides trigger an inflammatory response via Toll-like receptor 4 and promote the secretion of pro-inflammatory cytokines in the liver and adipose tissue [90,91]. This is known to maintain chronic inflammation in adipose tissue and exacerbate insulin resistance. It has also been suggested that reduced diversity of the intestinal microbiota leads to decreased short-chain fatty acid production and impaired intestinal barrier function, further contributing to inflammation [92].

3.2.6. Dermatological Diseases

Plant-derived compounds have attracted attention as promising natural ingredients for the prevention and treatment of skin diseases due to their diverse biological activities, including anti-inflammatory, antioxidant, and antibacterial effects. Phytoconstituents such as flavonoids, carotenoids, polyphenols, and terpenoids have been reported to have multifaceted effects on the major pathological conditions of skin diseases, such as skin inflammation, oxidative stress, and microbial infection [93,94].
For example, quercetin, luteolin, and apigenin, which are flavonoids, have been shown to be effective in alleviating inflammatory skin diseases such as atopic dermatitis and psoriasis by inhibiting the production of inflammatory cytokines such as TNF-α and IL-6 [95,96]. Quercetin contributes to the reduction of erythema and itching by inhibiting inflammatory signaling [96]. Carotenoids such as lycopene, lutein, and β-carotene have antioxidant properties, which include scavenging UV-induced ROS, and are useful in preventing photoaging and sun-induced skin damage. Oral lutein supplementation has been clinically shown to improve skin hydration and elasticity [97]. Polyphenols (e.g., resveratrol, EGCG, and chlorogenic acid) contribute to the control of acne and skin infections by enhancing skin barrier function and antibacterial activity. In particular, EGCG from green tea has been shown to inhibit sebum secretion and the growth of Cutibacterium acnes, suggesting its potential as a therapeutic aid for acne [94]. Terpenoids (curcumin, boswellic acid, cinnamaldehyde, and so on) exhibit anti-inflammatory effects as well as promote wound healing and antifungal activity. This has led to potential applications in contact dermatitis and fungal skin diseases. In addition, plant extracts and essential oils such as aloe vera, tea tree oil, calendula, and lavender oil are widely used to manage minor burns, itching, and irritating dermatitis due to their moisturizing, antibacterial, and soothing effects.
However, natural ingredients may cause allergic reactions or contact dermatitis in some individuals. Thus, it is important to evaluate safety and tolerability. In addition, challenges exist in the dermal application of plant-derived compounds, such as variability of ingredients, limitations in transdermal absorption, and lack of clinical evidence. To overcome these challenges, recent efforts have been made to improve skin penetration using nanocarrier technology and to develop standardized formulations.

4. Major Plant-Derived Compounds and Their Anti-Inflammatory Effects

As shown in Table 1, many plant-derived compounds are gaining attention in the prevention and treatment of chronic diseases because of their naturally occurring yet potent anti-inflammatory properties. These compounds exert their effects through the modulation of inflammatory cytokines, reduction in oxidative stress, and even modification of the immune response. Typical plant-derived compounds include polyphenols, flavonoids, carotenoids, terpenoids, and certain plant-derived molecules such as curcumin and gingerol.

4.1. Polyphenols

Polyphenols are secondary metabolites widely found in the plant kingdom and are known for their antioxidant and anti-inflammatory properties [98,99]. There are many different types of polyphenols, including flavonoids, phenolic acids, stilbenes, and lignans, among which the following examples are of particular interest:
As shown in Figure 2A, Quercetin is a type of flavonoid found mainly in apples and onions, with strong antioxidant and anti-inflammatory properties [100]. Resveratrol, on the other hand, is a stilbene compound found in red wine and grape skins and has been reported to have cardiovascular-protective and anti-inflammatory effects [101]. Furthermore, epigallocatechin gallate, which is found in green tea, has one of the strongest antioxidant activities among catechins and has been shown to inhibit inflammation [102]. These plant-derived compounds are expected to regulate inflammation through different mechanisms of action and contribute to reducing the risk of chronic diseases.
Polyphenols regulate inflammation through several key molecular mechanisms to exert their potent anti-inflammatory effects. First, polyphenols inhibit the activity of NF-κB [103]. NF-κB is a major transcription factor that regulates the expression of inflammatory cytokines, and suppression of its activation prevents an exaggerated inflammatory response. Second, polyphenols act as ROS scavengers and reduce intracellular oxidative stress [104]. Oxidative stress is an aggravating factor in inflammation, and its suppression plays an important role in the control of chronic inflammation. In addition, polyphenols reduce the progression of inflammation by inhibiting the production of inflammatory cytokines such as IL-1β, IL-6, and TNF-α [105]. Through these actions, polyphenols are expected to contribute to the suppression of chronic inflammation and the prevention of associated chronic diseases.
The anti-inflammatory effects of polyphenols have been demonstrated in various disease models and may contribute to preventing or inhibiting the progression of diseases related to chronic inflammation. For example, resveratrol has been shown to reduce the risk of CVD in animal models of the disease [106]. In particular, in atherosclerosis models, resveratrol was shown to reduce inflammation in vascular endothelial cells and prevent atherogenesis. This suggests that it may reduce the onset and progression of CVD. Quercetin has also been reported to reduce intestinal tissue damage in inflammatory bowel disease models by decreasing the expression of the inflammatory enzyme cyclooxygenase-2 [107]. This may improve symptoms and prevent inflammatory bowel disease. Furthermore, epigallocatechin gallate derived from green tea has been shown to suppress the production of inflammatory mediators secreted by fat cells in studies that involved obese mouse models (Figure 2B) [82]. This action has been suggested to reduce chronic inflammation, leading to a lower risk of diabetes through improved insulin resistance. These results indicate that polyphenols are promising candidates for the prevention and treatment of inflammatory diseases and warrant further study.
Flavonoids are a type of polyphenol, a group of compounds that give plants their pigments and bitter taste. Recent studies have revealed that flavonoids are more than just antioxidants; they selectively inhibit specific inflammatory pathways, thereby reducing chronic inflammation and likely contributing to the prevention and treatment of various diseases [108]. As a result, flavonoids are attracting attention as a new therapeutic strategy for inflammatory diseases. Luteolin and apigenin are representative flavonoids [109,110]. As shown in Figure 2C, Luteolin is abundant in celery and bell peppers and is known for its powerful antioxidant and anti-inflammatory properties [109]. Apigenin, on the other hand, is abundant in parsley and chamomile and has been reported to have anti-tumor and anti-inflammatory effects [110]. These flavonoids can inhibit inflammation and reduce disease progression through specific molecular mechanisms.
Flavonoids are known to inhibit the activity of the inflammatory enzyme cyclooxygenase-2 and induce nitric oxide synthase [111]. Induced nitric oxide synthase causes cellular damage by producing excessive nitric oxide. Excessive activity of these enzymes results in a persistent chronic inflammatory state, leading to tissue damage and disease progression. By decreasing the expression and activity of these enzymes, flavonoids may control the progression of inflammation and reduce the risk of developing chronic diseases [108]. Flavonoids are known to inhibit MAPK and NF-κB pathways, which are major signal transduction pathways in inflammation [112]. The MAPK pathway regulates the inflammatory response of cells to external stimuli and stress, and the NF-κB pathway is an important transcription factor that promotes the expression of inflammatory cytokines (IL-1β, IL-6, TNF-α, etc.) [113]. By inhibiting these pathways, flavonoids are considered to suppress inflammatory responses and prevent the vicious cycle of chronic inflammation. In particular, luteolin and apigenin have been reported to show high inhibitory activity against these pathways and effectively suppress inflammation [114].
It has been suggested that flavonoids may interact with the gut microbiota and potentiate their anti-inflammatory effects [115]. Gut bacteria are known to metabolize flavonoids and produce bioactive metabolites, and this metabolic process enhances the anti-inflammatory effects of flavonoids. Flavonoids may also reduce systemic inflammation levels by improving the balance of the gut microbiota and strengthening the gut barrier function [116]. Since intestinal bacterial imbalance plays a role in the progression of chronic diseases such as inflammatory bowel disease and metabolic syndrome, the effects of flavonoids on the intestinal environment are important from a disease prevention perspective.

4.2. Carotenoids

Carotenoids are fat-soluble pigments widely distributed in plants and algae that play an important role in maintaining human health. In particular, they are noted for their ability to protect cells from oxidative stress and reduce the risk of developing inflammatory diseases through their potent antioxidant properties [117]. Carotenoids are consumed in food and are known to provide various health benefits. Typical carotenoids include β-carotene (Figure 2D), lycopene (Figure 2E), and lutein (Figure 2F). β-carotene is abundant in carrots and squash and functions as a precursor to vitamin A in the body. Lycopene is abundant in tomatoes and watermelon and is known for its strong antioxidant properties, especially among carotenoids. Lutein, on the other hand, is found in spinach and kale and contributes primarily to the maintenance of eye health; it has also been reported to protect against inflammatory diseases.
Carotenoids protect cells from oxidative stress by inhibiting the oxidation (lipid peroxidation) of lipids that make up cell membranes [118]. Progressive lipid peroxidation leads to cell membrane damage and the activation of inflammatory responses, which increases the risk of developing chronic diseases [119]. Carotenoids help maintain tissue health by directly removing ROS and preventing lipid oxidation. Carotenoids reduce cellular oxidative stress by removing harmful ROS such as superoxide (O2-) and hydroxyl radicals (OH-) [120]. This prevents DNA and protein damage and inhibits the development of inflammation. Carotenoids also promote the activity of antioxidant enzymes [superoxide dismutase (SOD) and catalase] in the body and strengthen the endogenous antioxidant defense system [121].
Carotenoids modulate the balance of inflammation by promoting the production of anti-inflammatory cytokines (e.g., IL-10) while suppressing the production of inflammatory cytokines [122]. For example, lycopene and lutein have been reported to reduce chronic inflammation by suppressing the expression of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. This may reduce the progression of chronic diseases such as atherosclerosis and obesity-related diseases. Lutein has been shown to suppress the expression of inflammatory cytokines and reduce atherosclerotic lesions in mouse models of atherosclerosis [123]. Atherosclerosis progresses due to chronic inflammation of the vascular endothelium and is a major cause of CVD. Lutein’s anti-inflammatory action is expected to suppress the inflammation of the vascular endothelium and slow the progression of arteriosclerosis. In animal models of obesity, lycopene was shown to inhibit inflammatory pathways associated with liver fat accumulation [124]. Chronic inflammation associated with obesity is one of the factors that contribute to insulin resistance and non-alcoholic fatty liver disease [124]. Lycopene may reduce the risk of obesity-related diseases by reducing the expression of inflammatory mediators secreted by adipocytes and decreasing fat accumulation in the liver.

4.3. Curcumin and Gingerol

As shown in Figure 2G,H, curcumin and gingerol are bioactive compounds derived from plants and have long been used in traditional medicine. These compounds possess potent antioxidant and anti-inflammatory properties and have potential applications in the prevention and treatment of chronic diseases. In particular, by targeting inflammatory signaling pathways, these compounds may contribute to the amelioration of arthritis and chronic inflammatory diseases.
Curcumin is the yellow pigment in “turmeric” and is widely known for its antioxidant and anti-inflammatory properties [125]. Curcumin has traditionally been used in Indian Ayurvedic medicine and Chinese medicine, and in recent years, scientific research has revealed its diverse health benefits. Curcumin inhibits the NF-κB pathway, a major signaling pathway in inflammation, and suppresses the expression of inflammatory cytokines. NF-κB is a transcription factor that promotes the transcription of inflammatory cytokines such as IL-1β, IL-6, and TNF-α, which are involved in the development and progression of chronic inflammation. NF-κB is involved in the onset and progression of chronic inflammation [126]. Curcumin has been shown to reduce inflammatory responses and prevent tissue damage by inhibiting this pathway. Furthermore, curcumin has been reported to have inhibitory effects on cancer cell proliferation through the regulation of the cell cycle and induction of apoptosis (cell death) [127]. For this reason, research is being conducted on both its anti-inflammatory and anti-tumor effects. In animal experiments involving arthritis models, curcumin significantly reduced joint inflammation and alleviated joint pain [128]. Specifically, the administration of curcumin to mice models of arthritis reduced the expression of inflammatory cytokines and suppressed the destruction of joint tissue. Based on this, curcumin is believed to have potential as a therapeutic aid for rheumatoid arthritis and osteoarthritis.
Gingerol is the main bioactive constituent of “Ginger (Zingiber officinale)” and is also the substance responsible for ginger’s distinctive pungent taste. Gingerol has been reported to possess antioxidant and anti-inflammatory properties and is believed to be particularly effective in alleviating arthritis and digestive disorders [129]. Gingerol reduces chronic inflammation by inhibiting the secretion of TNF-α, a pro-inflammatory cytokine that is secreted by immune cells and plays a key role in amplifying inflammation and tissue destruction [130]. By suppressing the secretion of TNF-α, gingerol inhibits the progression of inflammation and contributes to the improvement of inflammation-related diseases. Gingerol has also been reported to reduce pain and swelling by inhibiting the activity of inflammatory enzymes (e.g., cyclooxygenase-2 and induced nitric oxide synthase) and suppressing the production of prostaglandins and nitric oxide [131]. In addition, gingerol suppresses oxidative stress responses and may contribute to the prevention of tissue damage. In mouse models of arthritis, gingerol was shown to inhibit the development of inflammation and reduce the production of inflammatory mediators [132]. Experiments have reported reduced joint inflammation and significantly decreased expression of inflammatory cytokines in mice treated with gingerol. Thus, gingerol is considered useful in relieving symptoms of rheumatoid arthritis and osteoarthritis.
Curcumin and gingerol have the advantage of being readily available as foods and supplements. Curcumin is found as turmeric in curry powder and other products, while gingerol is found in ginger-based foods and beverages. Daily consumption of these compounds may reduce the risk of chronic inflammation and prevent arthritis and other inflammatory diseases. However, curcumin is difficult to dissolve in water and has low bioavailability. For this reason, nanoparticulation and liposome technologies are being studied to enhance curcumin absorption. Gingerol is also being applied to the maintenance of digestive health and the prevention of metabolic diseases. In particular, research on gingerol’s effects on improving digestive function and anti-cancer activity is in progress, and further evidence is expected to be gathered through future clinical trials.

5. Molecular Mechanisms

A wide variety of molecular mechanisms are involved in the inhibition of inflammation by plant-derived compounds. In addition to modulating key inflammatory signaling pathways, these compounds regulate chronic inflammation by reducing oxidative stress and suppressing inflammatory gene expression.

5.1. Inhibition of the NF-κB Pathway

NF-κB is one of the major transcription factors that regulate the inflammatory response and regulates the expression of many inflammation-related genes, including inflammatory cytokines, chemokines, and adhesion molecules. In chronic inflammation, sustained activation of the NF-κB pathway is observed and plays a central role in many disease progressions. Plant-derived compounds exhibit potent anti-inflammatory effects through inhibition of the NF-κB pathway [133]. First, NF-κB normally exists in the cytoplasm bound to the inhibitory factor IκBα. However, when IκBα is phosphorylated and degraded by inflammatory stimuli (e.g., lipopolysaccharides and TNF-α), NF-κB moves into the nucleus and promotes transcription of inflammatory genes (IL-6, IL-8, TNF-α, etc.) [134]. Polyphenols (quercetin, resveratrol, etc.) inhibit the degradation of IκBα, thereby preventing nuclear transfer of NF-κB and suppressing inflammatory responses [135]. In addition, plant-derived compounds such as chalcone and apigenin directly bind to the p65 subunit of NF-κB and reduce its DNA binding ability, thereby preventing the progression of inflammation [136,137]. Furthermore, ROS are the main factors that activate the NF-κB pathway, and antioxidants such as epigallocatechin gallate eliminate ROS and indirectly inhibit NF-κB activation [138]. These mechanisms have been demonstrated in disease models, with curcumin dramatically reducing inflammatory cytokine production via inhibition of NF-κB in arthritis models and lycopene inhibiting the NF-κB pathway and reducing vascular endothelial inflammation in an atherosclerosis model.

5.2. Inhibition of the Inflammasome

The inflammasome is a cytosolic multiprotein complex that plays a pivotal role in the innate immune response by regulating the maturation and secretion of pro-inflammatory cytokines. Among various inflammasome types, the NLRP3 (NOD-like receptor family, pyrin domain-containing 3) inflammasome has been extensively studied due to its involvement in the pathogenesis of numerous chronic inflammatory conditions, including metabolic syndrome, neurodegenerative diseases, and autoimmune disorders [139]. Upon activation by a diverse range of stimuli—such as ROS, extracellular ATP, and crystalline substances—NLRP3 binds to the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) and pro-caspase-1, leading to the autocatalytic activation of caspase-1. This, in turn, cleaves pro-IL-1β and pro-IL-18 into their biologically active forms, IL-1β and IL-18, respectively, thereby amplifying the inflammatory response.
Emerging evidence highlights the regulatory role of plant-derived polyphenols and carotenoids in modulating NLRP3 inflammasome activation, offering a promising strategy for inflammation control and chronic disease prevention [140,141]. For instance, resveratrol and quercetin have been shown to attenuate IL-1β and IL-18 secretion by disrupting NLRP3 inflammasome assembly, likely through the inhibition of upstream signaling pathways such as NF-κB and ROS generation. These compounds may also stabilize mitochondrial membranes and preserve redox homeostasis, thereby reducing NLRP3 activation triggers.
Furthermore, luteolin suppresses NLRP3 activity by inhibiting the oligomerization of ASC, which is essential for inflammasome complex formation. This structural interference directly impairs the scaffold necessary for caspase-1 recruitment and activation, thus reducing downstream cytokine maturation. Carotenoids like lutein and lycopene exert similar anti-inflammatory effects through caspase-1 inhibition, which has been linked to decreased IL-1β production in models of metabolic syndrome and atherosclerosis.
These anti-inflammatory mechanisms have been substantiated in various disease models. EGCG, a major catechin in green tea, mitigates insulin resistance in type 2 diabetes models by downregulating NLRP3 inflammasome signaling. Similarly, curcumin has demonstrated neuroprotective properties in Alzheimer’s disease models by attenuating microglial activation and suppressing NLRP3-mediated neuroinflammation [142]. Such findings suggest that targeting the NLRP3 inflammasome with plant-derived compounds holds substantial potential for the development of nutraceuticals and adjunctive therapies for chronic inflammatory diseases.

5.3. Control of ROS Generation

Oxidative stress is a major factor that induces and exacerbates inflammation, and excessive production of ROS activates inflammatory signaling pathways. Through their antioxidant activity, plant-derived compounds inhibit ROS production and mitigate the progression of inflammation.
Plant-derived compounds reduce oxidative stress and inhibit the progression of inflammation through the induction of antioxidant enzymes and direct scavenging of ROS [143]. Polyphenols and flavonoids have been reported to reduce oxidative stress by increasing the expression of SOD and converting superoxide anions to hydrogen peroxide, while curcumin enhances SOD activity and inhibits oxidative stress-related inflammation in animal models. Resveratrol is also reported to be an effective antioxidant in the treatment of GPP [144]. Resveratrol also enhances glutathione peroxidase activity and prevents oxidative damage to cells by detoxifying hydrogen peroxide [145]. In addition, lycopene and β-carotene have free radical scavenging properties and protect cell function by preventing lipid oxidation of cell membranes [146]. In disease models, lutein has been shown to inhibit ROS formation in the liver of mice on a high-fat diet model and prevent the progression of fatty liver, while apigenin reduces oxidative stress in neurons and shows neuroprotective effects in a PD model [140]. Thus, plant-derived compounds target diverse molecular pathways and have great potential in the prevention and treatment of chronic diseases.

6. Challenges to Practical Application

To maximize the efficacy of plant-derived compounds and promote their practical application, the following issues must be resolved.

6.1. Increased Bioavailability

Many plant-derived compounds have been reported to have limited efficacy in the actual human body, although their effects have been clearly confirmed in vitro and in animal models. The main reason for this is low bioavailability. Bioavailability refers to the percentage of ingested compounds that reach the bloodstream and exert their effects on target tissues and cells. Many plant-derived compounds are highly fat-soluble and poorly water-soluble, which limits their absorption from the intestinal tract. Many polyphenols and flavonoids, in particular, may be degraded by intestinal bacteria and enzymes during the digestive process, reducing their activity. In some cases, after a compound is absorbed, its activity is lost due to metabolism in the liver (modification by Phase I and Phase II enzymes). Curcumin, for example, is known to be prone to a decrease in blood levels due to its rapid glucuronide conjugation. Some compounds may not be sufficiently effective against CNS diseases (e.g., AD) because they have difficulty crossing the blood–brain barrier. In the improvement of drug delivery systems, nanocarrier technology and delivery systems utilizing liposomes are attracting attention to improve bioavailability, and encapsulation of compounds in nanoparticles can improve intestinal absorption and control distribution to target tissues. Furthermore, some flavonoids and polyphenols are known to have bioactive metabolites by intestinal bacteria, and their combination with probiotics and prebiotics, which utilize this property to regulate the intestinal microflora, is promising.

6.2. Evaluation of Long-Term Safety

Plant-derived compounds are often considered safe because they are “naturally derived”, but comprehensive research on the risks and side effects of long-term ingestion is still limited. Potential risks associated with the safety of plant-derived compounds include accumulation toxicity, interaction problems, and allergic reactions. Some compounds that accumulate in the body with long-term ingestion may cause unexpected toxicity and side effects, particularly high concentrations of polyphenols, which have been reported to reverse antioxidant activity and induce oxidative stress. There is also concern about interaction issues, as these compounds can affect the metabolism of pharmaceuticals by inhibiting or inducing CYP450 enzymes, potentially altering drug efficacy and side effects. In addition, foods and supplements containing multiple polyphenols have been associated with the risk of allergic reactions in some consumers. To mitigate these risks, it is essential to conduct clinical trials, including long-term randomized controlled trials to assess safety, as well as comprehensive toxicity evaluations using animal models and cell studies.

6.3. Prospects for Practical Application

To overcome the challenges and effectively utilize plant-derived compounds in medicine and health promotion, the following innovations and new approaches are expected:
Nanocarrier technologies show promise for improving the absorption and target specificity of plant-derived compounds. Advanced drug delivery technologies such as liposomes, nanoemulsions, and polymer-based nanoparticles are being utilized to improve the bioavailability of plant-derived compounds. Liposomes encapsulate compounds in a lipid bilayer to improve absorption and allow efficient uptake from the intestinal tract, and liposomal formulations of curcumin, in particular, have been shown to increase bioavailability several-fold compared to conventional formulations. Nanoemulsions have attracted attention as a technology that converts poorly water-soluble compounds into nano-sized emulsified particles, improving their stability and solubility in the body. In addition, polymer-based nanoparticles using biodegradable polymers (e.g., PLGA) may provide sustained-release drug delivery by encapsulating plant-derived compounds, resulting in sustained efficacy.

6.4. Integration into Personalized Nutrition

In recent years, the field of nutrition has focused on “personalized nutrition” based on an individual’s genetic information and gut microbiota profile [147]. Incorporating this approach into the application of plant-derived compounds is expected to bring further benefits. In order to maximize the health benefits of plant-derived compounds, personalized intake planning and digital health technologies are being integrated [147]. In personalized intake planning, a system has been developed to suggest optimal compounds and intakes based on genetic information and the state of the intestinal microflora. In addition, through integration with digital health technology, a system is being built to monitor the effects of the intake of plant-derived compounds in real time using wearable devices and health management applications and is expected to establish optimal intake strategies based on individual health conditions [148].

6.5. Plant-Derived Dietary Fiber Resources and Their Effective Utilization

Plant-derived products contain a wide variety of dietary fibers, which are not only important for human health but are useful in the food industry [73,149,150]. In particular, in recent years, dietary fiber contained in inedible parts of fruits and vegetables (peels, seeds, cores, and so on) and edible by-products that tend to be discarded have been reevaluated, attracting attention from the perspective of reducing food loss in the agricultural food supply chain.
For example, apple lees, carrot and beet peels, cabbage leaves, and citrus peels and endosperm are rich in useful dietary fibers such as pectin, cellulose, and hemicellulose [151]. These dietary fibers have been reported to have various health functions, including intestinal regulation, blood sugar and lipid regulation, and even anti-inflammatory effects through the improvement of the intestinal microflora [152]. Furthermore, these by-product-derived fibers are expected to be utilized as functional foods, supplements, and alternative food materials (e.g., gluten-free products and texture modifiers) after powdering or extracting them. From the viewpoint of contributing to the construction of a sustainable food system, effective utilization of these plant-derived fibers is an important issue for future applied research.

7. Conclusions

Plant-derived compounds are of great interest in modern medicine and nutrition due to their diverse anti-inflammatory and chronic disease prevention potential. These compounds are abundant in nature and are chemically diverse, including polyphenols, flavonoids, carotenoids, and terpenoids. The potent antioxidant properties they exhibit and their ability to inhibit inflammatory cytokines, block inflammatory pathways, and even regulate the gut microbiota have been shown to play an important role in the prevention and inhibition of the progression of chronic diseases.
The greatest advantage of plant-derived compounds is their ability to approach the multifaceted drivers of chronic disease through their diverse biological effects. By targeting multiple pathways, such as NF-κB and the NLRP3 inflammasome, they exert anti-inflammatory and antioxidant effects, contributing to the prevention of cardiovascular and neurodegenerative diseases. In addition, plant-derived compounds regulate the gut microbiota and may affect mental health via the gut–brain axis, making them a potential complementary therapy for depression and anxiety disorders. The challenges for practical application of plant-derived compounds include improving bioavailability, establishing long-term safety, standardization and quality control, and overcoming legal regulations. Absorption must be improved through the use of nano-carrier technology and liposomes, and side effects and accumulated toxicity must be verified in long-term clinical trials. In addition, standardization must be promoted to prevent variations in quality, and appropriate legislation and scientific rationale must be strengthened for food and pharmaceutical products.

8. Future Directions

To advance applied research on plant-derived compounds, it is important to move in the direction of further elucidation of molecular mechanisms, expansion of clinical trials, integration with personalized nutrition, and collaboration with multiple disciplines. First, molecular elucidation of the anti-inflammatory and disease-preventive effects of plant-derived compounds, especially the multi-targeted mechanism of action, will enable more specific applications. However, it is essential to clarify the boundaries of their efficacy, as these compounds do not have the same benefits and individuals do not respond to them equally. Some plant-derived compounds may demonstrate promising effects in vitro or in animal models but fail to show significant clinical benefits in humans due to differences in bioavailability, metabolism, or dosage thresholds.
In addition, since much of the current research is based on animal models and cell studies, human clinical trials must be expanded and improved to confirm actual health effects and improve reliability. These trials should also aim to identify subpopulations that may benefit less—or not at all—from specific compounds, thus preventing overgeneralization of their efficacy. Furthermore, integration with personalized nutrition utilizing genetic information and gut microbiota profiles is expected to develop intake plans for plant-derived compounds that are optimized for individual patients. Additionally, collaboration among experts from multiple disciplines, including medicine, pharmacy, food science, and agriculture, will expand the range of applications of plant-derived compounds and promote their practical use in a wider range of fields.

Author Contributions

Conceptualization, methodology, writing—original draft preparation, writing—review and editing, K.N., N.I., K.K., and N.Y.; funding acquisition, K.N. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by JSPS KAKENHI Grant Number 23K08620 (K.N.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used and/or analyzed during this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CVDcardiovascular disease
IL-1βinterleukin-1β
IL-6interleukin-6
TNF-αtumor necrosis factor-α
ROSreactive oxygen species
NSAIDsnonsteroidal anti-inflammatory drugs
NF-κBnuclear factor-κB
MAPKmitogen-activated protein kinase
ADAlzheimer’s disease
PDParkinson’s disease
CNScentral nervous system
SODsuperoxide dismutase

References

  1. Papikinou, M.A.; Pavlidis, K.; Cholidis, P.; Kranas, D.; Adamantidi, T.; Anastasiadou, C.; Tsoupras, A. Marine Fungi Bioactives with Anti-Inflammatory, Antithrombotic and Antioxidant Health-Promoting Properties Against Inflammation-Related Chronic Diseases. Mar. Drugs 2024, 22, 520. [Google Scholar] [CrossRef]
  2. Pan, X.; Lv, J.; Liu, M.; Li, Y.; Zhang, Y.; Zhang, R.; Liu, J.; Sun, C.; Guo, H. Chronic systemic inflammation predicts long-term mortality among patients with fatty liver disease: Data from the National Health and Nutrition Examination Survey 2007–2018. PLoS ONE 2024, 19, e0312877. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, L.; Xu, Y.; Jiang, M.; Wang, M.; Ji, M.; Xie, X.; Sheng, H. Chronic stress induces depression-like behavior in rats through affecting brain mitochondrial function and inflammation. Psychoneuroendocrinology 2024, 172, 107261. [Google Scholar] [CrossRef]
  4. Martinez Nieto, M.; De Leon Rodriguez, M.L.; Anaya Macias, R.D.C.; Lomeli Martinez, S.M. Periodontitis and chronic kidney disease: A bidirectional relationship based on inflammation and oxidative stress. World J. Clin. Cases 2024, 12, 6775–6781. [Google Scholar] [CrossRef] [PubMed]
  5. Hertis Petek, T.; Marcun Varda, N. Childhood Cardiovascular Health, Obesity, and Some Related Disorders: Insights into Chronic Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2024, 25, 9706. [Google Scholar] [CrossRef] [PubMed]
  6. Brzozowski, T.; Konturek, P.; Konturek, S.J.; Kwiecien, S.; Sliwowski, Z.; Pajdo, R.; Duda, A.; Ptak, A.; Hahn, E.G. Implications of reactive oxygen species and cytokines in gastroprotection against stress-induced gastric damage by nitric oxide releasing aspirin. Int. J. Color. Dis. 2003, 18, 320–329. [Google Scholar] [CrossRef]
  7. MohanKumar, S.M.J.; Murugan, A.; Palaniyappan, A.; MohanKumar, P.S. Role of cytokines and reactive oxygen species in brain aging. Mech. Ageing Dev. 2023, 214, 111855. [Google Scholar] [CrossRef]
  8. Llanos, P.; Palomero, J. Reactive Oxygen and Nitrogen Species (RONS) and Cytokines-Myokines Involved in Glucose Uptake and Insulin Resistance in Skeletal Muscle. Cells 2022, 11, 4008. [Google Scholar] [CrossRef]
  9. Gonzalez, A.L.; Dungan, M.M.; Smart, C.D.; Madhur, M.S.; Doran, A.C. Inflammation Resolution in the Cardiovascular System: Arterial Hypertension, Atherosclerosis, and Ischemic Heart Disease. Antioxid. Redox Signal 2024, 40, 292–316. [Google Scholar] [CrossRef]
  10. Shimabukuro, M. IGF-1 and Cardiovascular and Non-Cardiovascular Mortality Risk in Patients with Chronic Kidney Disease: A Model of “Malnutrition-Inflammation-Atherosclerosis Syndrome”. J. Atheroscler. Thromb. 2022, 29, 1138–1139. [Google Scholar] [CrossRef]
  11. Hong, S.; Dimitrov, S.; Cheng, T.; Redwine, L.; Pruitt, C.; Mills, P.J.; Ziegler, M.G.; Green, J.M.; Shaikh, F.; Wilson, K. Beta-adrenergic receptor mediated inflammation control by monocytes is associated with blood pressure and risk factors for cardiovascular disease. Brain Behav. Immun. 2015, 50, 31–38. [Google Scholar] [CrossRef] [PubMed]
  12. Conen, D. Inflammation, blood pressure and cardiovascular disease: Heading east. J. Hum. Hypertens 2013, 27, 71. [Google Scholar] [CrossRef]
  13. Curro, D.; Vergani, E.; Bruno, C.; Comi, S.; D’Abate, C.; Mancini, A. Plasmatic lipocalin-2 levels in chronic low-grade inflammation syndromes: Comparison between metabolic syndrome, total and partial adult growth hormone deficiency. Biofactors 2020, 46, 629–636. [Google Scholar] [CrossRef]
  14. Rumyantsev, K.A.; Polyakova, V.V.; Sorokina, I.V.; Feoktistova, P.S.; Khatkov, I.E.; Bodunova, N.A.; Zhukova, L.G. The Gut Microbiota Impacts Gastrointestinal Cancers through Obesity, Diabetes, and Chronic Inflammation. Life 2024, 14, 1219. [Google Scholar] [CrossRef]
  15. Zhai, W.; Zhao, M.; Wen, C.; Meng, L.; Zhao, A.; Zhang, Y.; Cui, X.; Xu, Y.; Sun, L. Obesity-induced chronic low-grade inflammation in adipose tissue: A pathway to Alzheimer’s disease. Ageing Res. Rev. 2024, 99, 102402. [Google Scholar] [CrossRef]
  16. Subeta, P.; Lana, A.J.; Schlachetzki, J.C.M. Chronic peripheral inflammation: A possible contributor to neurodegenerative diseases. Neural Regen. Res. 2021, 16, 1711–1714. [Google Scholar] [CrossRef]
  17. Saeed, B.A.; Faisal, A.J.; Mahmood, B.S.; Thanoon, A.H. Chronic Inflammation Induced by Escherichia coli Blood Infections as a Risk Factor for Pancreatic Cancer Progression. Asian Pac. J. Cancer Prev. 2024, 25, 4407–4414. [Google Scholar] [CrossRef]
  18. Rogovskii, V. Cancer and Autoimmune Diseases as Two Sides of Chronic Inflammation and the Method of Therapy. Curr. Cancer Drug Targets 2024, 24, 1089–1103. [Google Scholar] [CrossRef] [PubMed]
  19. Zadori, Z.S.; Kiraly, K.; Al-Khrasani, M.; Gyires, K. Interactions between NSAIDs, opioids and the gut microbiota—Future perspectives in the management of inflammation and pain. Pharmacol. Ther. 2023, 241, 108327. [Google Scholar] [CrossRef]
  20. Han, Y.; Lu, Y.; Li, X.; Niu, X.; Chang, A.K.; Yang, Z.; Li, X.; He, X.; Bi, X. Novel organoselenides (NSAIDs-Se derivatives) protect against LPS-induced inflammation in microglia by targeting the NOX2/NLRP3 signaling pathway. Int. Immunopharmacol. 2021, 101, 108377. [Google Scholar] [CrossRef]
  21. Martin, C.S.; Crastin, A.; Sagmeister, M.S.; Kalirai, M.S.; Turner, J.D.; MacDonald, L.; Kurowska-Stolarska, M.; Scheel-Toellner, D.; Taylor, A.E.; Gilligan, L.C.; et al. Inflammation dynamically regulates steroid hormone metabolism and action within macrophages in rheumatoid arthritis. J. Autoimmun. 2024, 147, 103263. [Google Scholar] [CrossRef] [PubMed]
  22. Ardelean, M.; Buzas, R.; Ardelean, O.; Preda, M.; Morariu, S.I.; Levai, C.M.; Rosca, C.I.; Lighezan, D.F.; Kundnani, N.R. Clinical and Biochemical Differences in Patients Having Non-Variceal Upper Gastrointestinal Bleeding on NSAIDs, Oral Anticoagulants, and Antiplatelet Therapy. J. Clin. Med. 2024, 13, 5622. [Google Scholar] [CrossRef]
  23. Zhang, W.; Xu, X.; Zhang, R.; Tian, Y.; Ma, X.; Wang, X.; Jiang, Y.; Man, C. Stress-Induced Immunosuppression Inhibits Regional Immune Responses in Chicken Adipose Tissue Partially through Suppressing T Cells by Up-Regulating Steroid Metabolism. Animals 2024, 14, 225. [Google Scholar] [CrossRef]
  24. Baron, G.; Ferrario, G.; Marinello, C.; Carini, M.; Morazzoni, P.; Aldini, G. Effect of Extraction Solvent and Temperature on Polyphenol Profiles, Antioxidant and Anti-Inflammatory Effects of Red Grape Skin By-Product. Molecules 2021, 26, 5454. [Google Scholar] [CrossRef]
  25. Wu, H.; Pang, H.; Chen, Y.; Huang, L.; Liu, H.; Zheng, Y.; Sun, C.; Zhang, G.; Wang, G. Anti-Inflammatory Effect of a Polyphenol-Enriched Fraction from Acalypha wilkesiana on Lipopolysaccharide-Stimulated RAW 264.7 Macrophages and Acetaminophen-Induced Liver Injury in Mice. Oxid. Med. Cell Longev. 2018, 2018, 7858094. [Google Scholar] [CrossRef] [PubMed]
  26. Cirmi, S.; Randazzo, B.; Russo, C.; Musumeci, L.; Maugeri, A.; Montalbano, G.; Guerrera, M.C.; Lombardo, G.E.; Levanti, M. Anti-inflammatory effect of a flavonoid-rich extract of orange juice in adult zebrafish subjected to Vibrio anguillarum-induced enteritis. Nat. Prod. Res. 2021, 35, 5350–5353. [Google Scholar] [CrossRef]
  27. Unal, N.G.; Kozak, A.; Karakaya, S.; Oruc, N.; Barutcuoglu, B.; Aktan, C.; Sezak, M.; Ozutemiz, A.O. Anti-Inflammatory Effect of Crude Momordica charantia L. Extract on 2,4,6-Trinitrobenzene Sulfonic Acid-Induced Colitis Model in Rat and the Bioaccessibility of its Carotenoid Content. J. Med. Food 2020, 23, 641–648. [Google Scholar] [CrossRef] [PubMed]
  28. Mohammad, C.A.; Ali, K.M.; Sha, A.M.; Gul, S.S. Effect of Curcumin gel on inflammatory and anti-inflammatory biomarkers in experimental induced periodontitis in rats: A biochemical and immunological study. Front. Microbiol. 2023, 14, 1274189. [Google Scholar] [CrossRef] [PubMed]
  29. Song, S.; Dang, M.; Kumar, M. Anti-inflammatory and renal protective effect of gingerol in high-fat diet/streptozotocin-induced diabetic rats via inflammatory mechanism. Inflammopharmacology 2019, 27, 1243–1254. [Google Scholar] [CrossRef]
  30. Li, Z.; Liu, T.; Xie, W.; Wang, Z.; Gong, B.; Yang, M.; He, Y.; Bai, X.; Liu, K.; Xie, Z.; et al. Protopanaxadiol derivative: A plant origin of novel selective glucocorticoid receptor modulator with anti-inflammatory effect. Eur. J. Pharmacol. 2024, 983, 176901. [Google Scholar] [CrossRef]
  31. Faubel, N.; Makran, M.; Barbera, R.; Garcia-Llatas, G.; Giardina, I.C.; Tesoriere, L.; Attanzio, A.; Cilla, A. Anti-inflammatory activity of plant sterols in a co-culture model of intestinal inflammation: Focus on food-matrix effect. Food Funct. 2024, 15, 6502–6511. [Google Scholar] [CrossRef] [PubMed]
  32. Ma, A.; Zhou, J.; Zou, H.; Yuan, L.; Zhong, R.; Zhu, Y.; Gao, C. Anti-inflammatory effect of nestorone in a lipopolysaccharide-induced acute lung injury model through regulation of the TLR-4/Myd88/NF-kappaB signaling pathway. Inflammopharmacology 2024, 33, 1473–1489. [Google Scholar] [CrossRef] [PubMed]
  33. Hao, R.; Liu, Q.; Wang, L.; Jian, W.; Cheng, Y.; Zhang, Q.; Hayer, K.; Kamarudin Raja Idris, R.; Zhang, Y.; Lu, H.; et al. Anti-inflammatory effect of Lactiplantibacillus plantarum T1 cell-free supernatants through suppression of oxidative stress and NF-kappaB- and MAPK-signaling pathways. Appl. Environ. Microbiol. 2023, 89, e0060823. [Google Scholar] [CrossRef]
  34. Taher, I.; El-Masry, E.; Abouelkheir, M.; Taha, A.E. Anti-inflammatory effect of metformin against an experimental model of LPS-induced cytokine storm. Exp. Ther. Med. 2023, 26, 415. [Google Scholar] [CrossRef] [PubMed]
  35. Jung, M.A.; Jang, S.E.; Hong, S.W.; Hana, M.J.; Kim, D.H. The role of intestinal microflora in anti-inflammatory effect of baicalin in mice. Biomol. Ther. 2012, 20, 36–42. [Google Scholar] [CrossRef]
  36. Mu, W.; Patankar, V.; Kitchen, S.; Zhen, A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses 2024, 16, 219. [Google Scholar] [CrossRef]
  37. Winston, C.N.; Noel, A.; Neustadtl, A.; Parsadanian, M.; Barton, D.J.; Chellappa, D.; Wilkins, T.E.; Alikhani, A.D.; Zapple, D.N.; Villapol, S.; et al. Dendritic Spine Loss and Chronic White Matter Inflammation in a Mouse Model of Highly Repetitive Head Trauma. Am. J. Pathol. 2016, 186, 552–567. [Google Scholar] [CrossRef]
  38. Corrado, A.; Guadagni, I.; Picarelli, G.; Variola, A. Obesity and Chronic Inflammation: Implications for Rheumatoid Arthritis, Spondyloarthritis, and Ulcerative Colitis. Immun. Inflamm. Dis. 2025, 13, e70080. [Google Scholar] [CrossRef]
  39. Kurochkina, N.S.; Orlova, M.A.; Vigovskiy, M.A.; Zgoda, V.G.; Vepkhvadze, T.F.; Vavilov, N.E.; Makhnovskii, P.A.; Grigorieva, O.A.; Boroday, Y.R.; Philippov, V.V.; et al. Age-related changes in human skeletal muscle transcriptome and proteome are more affected by chronic inflammation and physical inactivity than primary aging. Aging Cell 2024, 23, e14098. [Google Scholar] [CrossRef]
  40. Yang, Y.; Wang, H.; Wang, X.; Chen, L.; Liu, W.; Cai, D.; Deng, S.; Chu, H.; Liu, Y.; Feng, X.; et al. Long-term environmental levels of microcystin-LR exposure induces colorectal chronic inflammation, fibrosis and barrier disruption via CSF1R/Rap1b signaling pathway. J. Hazard. Mater. 2022, 440, 129793, Corrigendum in J. Hazard. Mater. 2023, 451, 131110. [Google Scholar] [CrossRef]
  41. Berger, M.; Maqua, H.; Lysaja, K.; Mause, S.F.; Hindle, M.S.; Naseem, K.; Dahl, E.; Speer, T.; Marx, N.; Schutt, K. Platelets from patients with chronic inflammation have a phenotype of chronic IL-1beta release. Res. Pract. Thromb. Haemost. 2024, 8, 102261. [Google Scholar] [CrossRef] [PubMed]
  42. Sater, M.S.; AlDehaini, D.M.B.; Malalla, Z.H.A.; Ali, M.E.; Giha, H.A. Plasma IL-6, TREM1, uPAR, and IL6/IL8 biomarkers increment further witnessing the chronic inflammation in type 2 diabetes. Horm. Mol. Biol. Clin. Investig. 2023, 44, 259–269. [Google Scholar] [CrossRef]
  43. Joveini, S.; Yarmohammadi, F.; Iranshahi, M.; Nikpoor, A.R.; Askari, V.R.; Attaranzadeh, A.; Etemad, L.; Taherzadeh, Z. Distinct therapeutic effects of auraptene and umbelliprenin on TNF-alpha and IL-17 levels in a murine model of chronic inflammation. Heliyon 2024, 10, e40731. [Google Scholar] [CrossRef] [PubMed]
  44. Perez-Figueroa, E.; Alvarez-Carrasco, P.; Ortega, E. Crosslinking of membrane CD13 in human neutrophils mediates phagocytosis and production of reactive oxygen species, neutrophil extracellular traps and proinflammatory cytokines. Front. Immunol. 2022, 13, 994496. [Google Scholar] [CrossRef] [PubMed]
  45. Dong, P.; Qiu, H.; Wen, R.; Zou, X.; Sun, X.; Yu, L.; Zhang, S.; Wu, Y.; Lan, F. Reactive Oxygen and Nitrogen Species—“Nanosweeper” for Rheumatoid Arthritis Theranostics by Macrophage Reprogramming. ACS Appl. Mater. Interfaces 2024, 16, 70322–70338. [Google Scholar] [CrossRef]
  46. Ma, E.; Sasazuki, S.; Shimazu, T.; Sawada, N.; Yamaji, T.; Iwasaki, M.; Inoue, M.; Tsugane, S. Reactive oxygen species and gastric cancer risk: A large nested case-control study in Japan. Eur. J. Epidemiol. 2015, 30, 589–594. [Google Scholar] [CrossRef]
  47. Wang, Z.; Li, M.; Chen, J.; Zhang, S.; Wang, B.; Wang, J. Immunomodulatory Hydrogel for Electrostatically Capturing Pro-inflammatory Factors and Chemically Scavenging Reactive Oxygen Species in Chronic Diabetic Wound Remodeling. Adv. Heal. Mater. 2024, 13, e2402080. [Google Scholar] [CrossRef]
  48. Dughbaj, M.A.; Jayne, J.G.; Park, A.Y.J.; Bensman, T.J.; Algorri, M.; Ouellette, A.J.; Selsted, M.E.; Beringer, P.M. Anti-Inflammatory Effects of RTD-1 in a Murine Model of Chronic Pseudomonas aeruginosa Lung Infection: Inhibition of NF-kappaB, Inflammasome Gene Expression, and Pro-IL-1beta Biosynthesis. Antibiotics 2021, 10, 1043. [Google Scholar] [CrossRef]
  49. Nerland, D.; Ash, A.; Garman, A.; Foltz, J.; Berenbeim, G.; Wilke, B.; Winter, L.; Christian, D.T.; Duric, V. Chronic Inflammatory Pain Alters Expression of Limbic MAPK Phosphatases. Chronic Pain. Manag. 2024, 8, 155. [Google Scholar] [CrossRef]
  50. Suikkila, A.; Lyly, A.; Savinko, T.; Vento, S.I.; Saarinen, R.; Hafren, L. Inflammatory Cytokines in Middle Ear Effusion of Patients With Asthma, Chronic Rhinosinusitis With Nasal Polyps With or Without NSAID Intolerance. Otol. Neurotol. 2024, 45, 765–772. [Google Scholar] [CrossRef]
  51. Ajrawat, P.; Yang, Y.; Wasilewski, E.; Leroux, T.; Ladha, K.S.; Bhatia, A.; Singh, M.; Thaker, S.; Kapoor, M.; Furlan, A.D.; et al. Medical Cannabis Use and Inflammatory Cytokines and Chemokines Among Adult Chronic Pain Patients. Cannabis Cannabinoid Res. 2024, 9, 267–281. [Google Scholar] [CrossRef] [PubMed]
  52. Schmidt, C.; Baumeister, B.; Kipnowski, J.; Schiermeyer-Dunkhase, B.; Vetter, H. Alteration of prostaglandin E2 and leukotriene B4 synthesis in chronic inflammatory bowel disease. Hepatogastroenterology 1996, 43, 1508–1512. [Google Scholar]
  53. Kokkosis, A.G.; Madeira, M.M.; Hage, Z.; Valais, K.; Koliatsis, D.; Resutov, E.; Tsirka, S.E. Chronic psychosocial stress triggers microglial-/macrophage-induced inflammatory responses leading to neuronal dysfunction and depressive-related behavior. Glia 2024, 72, 111–132. [Google Scholar] [CrossRef]
  54. Huriyati, E.; Darwin, E.; Yanwirasti, Y.; Wahid, I. Differences in Expression of Inflammatory Mediator in Mucosal and Polyp Tissue between Chronic Rhinosinusitis and Recurrent Chronic Rhinosinusitis. Open Access Maced. J. Med. Sci. 2019, 7, 1733–1738. [Google Scholar] [CrossRef]
  55. Chai, B.; Qiao, Y.; Wang, H.; Zhang, X.; Wang, J.; Wang, C.; Zhou, P.; Chen, X. Identification of YfiH and the Catalase CatA As Polyphenol Oxidases of Aeromonas media and CatA as a Regulator of Pigmentation by Its Peroxyl Radical Scavenging Capacity. Front. Microbiol. 2017, 8, 1939. [Google Scholar] [CrossRef] [PubMed]
  56. Garcia-de Los Santos, A.; Lopez, E.; Cubillas, C.A.; Noel, K.D.; Brom, S.; Romero, D. Requirement of a plasmid-encoded catalase for survival of Rhizobium etli CFN42 in a polyphenol-rich environment. Appl. Environ. Microbiol. 2008, 74, 2398–2403. [Google Scholar] [CrossRef] [PubMed]
  57. Kim, J.Y.; Choi, J.Y.; Lee, H.J.; Byun, C.J.; Park, J.H.; Park, J.H.; Cho, H.S.; Cho, S.J.; Jo, S.A.; Jo, I. The Green Tea Component (-)-Epigallocatechin-3-Gallate Sensitizes Primary Endothelial Cells to Arsenite-Induced Apoptosis by Decreasing c-Jun N-Terminal Kinase-Mediated Catalase Activity. PLoS ONE 2015, 10, e0138590. [Google Scholar] [CrossRef]
  58. Chen, B.; He, Q.; Chen, C.; Lin, Y.; Xiao, J.; Pan, Z.; Li, M.; Li, S.; Yang, J.; Wang, F.; et al. Combination of curcumin and catalase protects against chondrocyte injury and knee osteoarthritis progression by suppressing oxidative stress. Biomed. Pharmacother. 2023, 168, 115751. [Google Scholar] [CrossRef]
  59. Qi, X.; Meng, J.; Li, C.; Cheng, W.; Fan, A.; Huang, J.; Lin, W. Praelolide alleviates collagen-induced arthritis through increasing catalase activity and activating Nrf2 pathway. Phytomedicine 2024, 135, 156040. [Google Scholar] [CrossRef]
  60. Oyedemi, S.O.; Nwaogu, G.; Chukwuma, C.I.; Adeyemi, O.T.; Matsabisa, M.G.; Swain, S.S.; Aiyegoro, O.A. Quercetin modulates hyperglycemia by improving the pancreatic antioxidant status and enzymes activities linked with glucose metabolism in type 2 diabetes model of rats: In silico studies of molecular interaction of quercetin with hexokinase and catalase. J. Food Biochem. 2020, 44, e13127. [Google Scholar] [CrossRef]
  61. Bensenor, I.M.; Goulart, A.C.; Pereira, A.C.; Brunoni, A.R.; Alencar, A.; Santos, R.D.; Bittencourt, M.S.; Telles, R.W.; Machado, L.A.C.; Barreto, S.M.; et al. Chronic inflammatory diseases, subclinical atherosclerosis, and cardiovascular diseases: Design, objectives, and baseline characteristics of a prospective case-cohort study—ELSA-Brasil. Clinics 2022, 77, 100013. [Google Scholar] [CrossRef] [PubMed]
  62. Hansen, P.R. Chronic Inflammatory Diseases and Atherosclerotic Cardiovascular Disease: Innocent Bystanders or Partners in Crime? Curr. Pharm. Des. 2018, 24, 281–290. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, H.W.; Guo, Y.L.; Wu, N.Q.; Zhu, C.G.; Dong, Q.; Sun, J.; Dou, K.F.; Li, J.J. Low-density lipoprotein triglyceride predicts outcomes in patients with chronic coronary syndrome following percutaneous coronary intervention according to inflammatory status. Clin. Chim. Acta 2023, 551, 117631. [Google Scholar] [CrossRef] [PubMed]
  64. Weissman, S.; Sinh, P.; Mehta, T.I.; Thaker, R.K.; Derman, A.; Heiberger, C.; Qureshi, N.; Amrutiya, V.; Atoot, A.; Dave, M.; et al. Atherosclerotic cardiovascular disease in inflammatory bowel disease: The role of chronic inflammation. World J. Gastrointest. Pathophysiol. 2020, 11, 104–113. [Google Scholar] [CrossRef]
  65. Straub, R.H. Insulin resistance, selfish brain, and selfish immune system: An evolutionarily positively selected program used in chronic inflammatory diseases. Arthritis Res. Ther. 2014, 16 (Suppl. S2), S4. [Google Scholar] [CrossRef]
  66. Li, D.; Wu, Y.; Tian, P.; Zhang, X.; Wang, H.; Wang, T.; Ying, B.; Wang, L.; Shen, Y.; Wen, F. Adipokine CTRP-5 as a Potential Novel Inflammatory Biomarker in Chronic Obstructive Pulmonary Disease. Medicine 2015, 94, e1503. [Google Scholar] [CrossRef]
  67. Liu, W.S.; Zhang, Y.R.; Ge, Y.J.; Wang, H.F.; Cheng, W.; Yu, J.T. Inflammation and Brain Structure in Alzheimer’s Disease and Other Neurodegenerative Disorders: A Mendelian Randomization Study. Mol. Neurobiol. 2024, 61, 1593–1604. [Google Scholar] [CrossRef]
  68. Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
  69. Mella, C.; Tsarouhas, P.; Brockwell, M.; Ball, H.C. The Role of Chronic Inflammation in Pediatric Cancer. Cancers 2025, 17, 154. [Google Scholar] [CrossRef]
  70. Guadagni, F.; Ferroni, P.; Palmirotta, R.; Portarena, I.; Formica, V.; Roselli, M. Review. TNF/VEGF cross-talk in chronic inflammation-related cancer initiation and progression: An early target in anticancer therapeutic strategy. In Vivo 2007, 21, 147–161. [Google Scholar]
  71. Lee, H.T.; Lin, C.S.; Liu, C.Y.; Chen, P.; Tsai, C.Y.; Wei, Y.H. Mitochondrial Plasticity and Glucose Metabolic Alterations in Human Cancer under Oxidative Stress-From Viewpoints of Chronic Inflammation and Neutrophil Extracellular Traps (NETs). Int. J. Mol. Sci. 2024, 25, 9458. [Google Scholar] [CrossRef]
  72. Bartsch, H.; Nair, J. Chronic inflammation and oxidative stress in the genesis and perpetuation of cancer: Role of lipid peroxidation, DNA damage, and repair. Langenbecks Arch. Surg. 2006, 391, 499–510. [Google Scholar] [CrossRef]
  73. Sorrentino, C.; Di Gisi, M.; Gentile, G.; Licitra, F.; D’Angiolo, R.; Giovannelli, P.; Migliaccio, A.; Castoria, G.; Di Donato, M. Agri-Food By-Products in Cancer: New Targets and Strategies. Cancers 2022, 14, 5517. [Google Scholar] [CrossRef]
  74. Yarden, Y.; Pines, G. The ERBB network: At last, cancer therapy meets systems biology. Nat. Rev. Cancer 2012, 12, 553–563. [Google Scholar] [CrossRef] [PubMed]
  75. Mohiuddin, M.; Kasahara, K. Cisplatin Activates the Growth Inhibitory Signaling Pathways by Enhancing the Production of Reactive Oxygen Species in Non-small Cell Lung Cancer Carrying an EGFR Exon 19 Deletion. Cancer Genom. Proteom. 2021, 18, 471–486. [Google Scholar] [CrossRef] [PubMed]
  76. Chakrabarti, M.; Ray, S.K. Direct transfection of miR-137 mimics is more effective than DNA demethylation of miR-137 promoter to augment anti-tumor mechanisms of delphinidin in human glioblastoma U87MG and LN18 cells. Gene 2015, 573, 141–152. [Google Scholar] [CrossRef]
  77. Zatterale, F.; Longo, M.; Naderi, J.; Raciti, G.A.; Desiderio, A.; Miele, C.; Beguinot, F. Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes. Front. Physiol. 2019, 10, 1607. [Google Scholar] [CrossRef] [PubMed]
  78. Rakotoarivelo, V.; Lacraz, G.; Mayhue, M.; Brown, C.; Rottembourg, D.; Fradette, J.; Ilangumaran, S.; Menendez, A.; Langlois, M.F.; Ramanathan, S. Inflammatory Cytokine Profiles in Visceral and Subcutaneous Adipose Tissues of Obese Patients Undergoing Bariatric Surgery Reveal Lack of Correlation With Obesity or Diabetes. EBioMedicine 2018, 30, 237–247. [Google Scholar] [CrossRef] [PubMed]
  79. Wei, D.; Tian, X.; Ren, Z.; Liu, Z.; Sun, C. Mechanistic insights into the role of USP14 in adipose tissue macrophage recruitment and insulin resistance in obesity. Int. J. Biol. Macromol. 2024, 267, 131645. [Google Scholar] [CrossRef]
  80. Nakadate, K.; Kawakami, K.; Yamazaki, N. Combined Ingestion of Tea Catechin and Citrus beta-Cryptoxanthin Improves Liver Function via Adipokines in Chronic Obesity. Nutrients 2023, 15, 3345. [Google Scholar] [CrossRef]
  81. Nakadate, K.; Kawakami, K.; Yamazaki, N. Anti-Obesity and Anti-Inflammatory Synergistic Effects of Green Tea Catechins and Citrus β-Cryptoxanthin Ingestion in Obese Mice. Int. J. Mol. Sci. 2023, 24, 7054. [Google Scholar] [CrossRef] [PubMed]
  82. Nakadate, K.; Kawakami, K.; Yamazaki, N. Synergistic Effect of beta-Cryptoxanthin and Epigallocatechin Gallate on Obesity Reduction. Nutrients 2024, 16, 2344. [Google Scholar] [CrossRef]
  83. Saitoh, H.; Sakaguchi, M.; Miruno, F.; Muramatsu, N.; Ito, N.; Tadokoro, K.; Kawakami, K.; Nakadate, K. Histopathological Analysis of Lipopolysaccharide-Induced Liver Inflammation and Thrombus Formation in Mice: The Protective Effects of Aspirin. Curr. Issues Mol. Biol. 2024, 46, 14291–14303. [Google Scholar] [CrossRef] [PubMed]
  84. Dousdampanis, P.; Aggeletopoulou, I.; Mouzaki, A. The role of M1/M2 macrophage polarization in the pathogenesis of obesity-related kidney disease and related pathologies. Front. Immunol. 2024, 15, 1534823. [Google Scholar] [CrossRef]
  85. Lee, C.H.; Kim, H.J.; Lee, Y.S.; Kang, G.M.; Lim, H.S.; Lee, S.H.; Song, D.K.; Kwon, O.; Hwang, I.; Son, M.; et al. Hypothalamic Macrophage Inducible Nitric Oxide Synthase Mediates Obesity-Associated Hypothalamic Inflammation. Cell Rep. 2018, 25, 934–946.e935. [Google Scholar] [CrossRef]
  86. Cao, X.; Gao, T.; Lv, F.; Wang, Y.; Li, B.; Wang, X. ROS-triggered and macrophage-targeted micelles modulate mitochondria function and polarization in obesity. Nanotechnology 2024, 35, 475707. [Google Scholar] [CrossRef]
  87. Wu, Z.; Yu, W.; Ni, W.; Teng, C.; Ye, W.; Yu, C.; Zeng, Y. Improvement of obesity by Liupao tea is through the IRS-1/PI3K/AKT/GLUT4 signaling pathway according to network pharmacology and experimental verification. Phytomedicine 2023, 110, 154633. [Google Scholar] [CrossRef]
  88. Feng, M.; Liu, F.; Xing, J.; Zhong, Y.; Zhou, X. Anemarrhena saponins attenuate insulin resistance in rats with high-fat diet-induced obesity via the IRS-1/PI3K/AKT pathway. J. Ethnopharmacol. 2021, 277, 114251. [Google Scholar] [CrossRef] [PubMed]
  89. Mao, X.; Paerhati, G.; Wu, Y.; Cheng, L.F. Modulation of gut microbiota, up-regulation of ZO-1, and promotion of metabolism as therapeutic mechanisms of indole-3-carbinol against obesity in mice. Front. Pharmacol. 2024, 15, 1499142. [Google Scholar] [CrossRef]
  90. Keirns, B.H.; Medlin, A.R.; Maki, K.A.; McClanahan, K.; Fruit, S.E.; Sciarrillo, C.M.; Hart, S.M.; Joyce, J.; Lucas, E.A.; Emerson, S.R. Biomarkers of intestinal permeability are associated with inflammation in metabolically healthy obesity but not normal-weight obesity. Am. J. Physiol. Heart Circ. Physiol. 2024, 327, H1135–H1145. [Google Scholar] [CrossRef]
  91. Wu, J.; Liu, Y.; Dou, Z.; Wu, T.; Liu, R.; Sui, W.; Jin, Y.; Zhang, M. Black garlic melanoidins prevent obesity, reduce serum LPS levels and modulate the gut microbiota composition in high-fat diet-induced obese C57BL/6J mice. Food Funct. 2020, 11, 9585–9598. [Google Scholar] [CrossRef] [PubMed]
  92. Kopczynska, J.; Kowalczyk, M. The potential of short-chain fatty acid epigenetic regulation in chronic low-grade inflammation and obesity. Front. Immunol. 2024, 15, 1380476. [Google Scholar] [CrossRef] [PubMed]
  93. Sharma, V.; Gautam, D.N.S.; Radu, A.F.; Behl, T.; Bungau, S.G.; Vesa, C.M. Reviewing the Traditional/Modern Uses, Phytochemistry, Essential Oils/Extracts and Pharmacology of Embelia ribes Burm. Antioxidants 2022, 11, 1359. [Google Scholar] [CrossRef]
  94. Khuntia, A.; Martorell, M.; Ilango, K.; Bungau, S.G.; Radu, A.F.; Behl, T.; Sharifi-Rad, J. Theoretical evaluation of Cleome species’ bioactive compounds and therapeutic potential: A literature review. Biomed. Pharmacother. 2022, 151, 113161. [Google Scholar] [CrossRef]
  95. Yang, S.C.; Chang, Z.Y.; Hsiao, C.Y.; Alshetaili, A.; Wei, S.H.; Hsiao, Y.T.; Fang, J.Y. Topical Anti-Inflammatory Effects of Quercetin Glycosides on Atopic Dermatitis-Like Lesions: Influence of the Glycone Type on Efficacy and Skin Absorption. Inflammation 2025. [Google Scholar] [CrossRef] [PubMed]
  96. Beken, B.; Serttas, R.; Yazicioglu, M.; Turkekul, K.; Erdogan, S. Quercetin Improves Inflammation, Oxidative Stress, and Impaired Wound Healing in Atopic Dermatitis Model of Human Keratinocytes. Pediatr. Allergy Immunol. Pulmonol. 2020, 33, 69–79. [Google Scholar] [CrossRef]
  97. Nolan, J.M.; Power, R.; Howard, A.N.; Bergin, P.; Roche, W.; Prado-Cabrero, A.; Pope, G.; Cooke, J.; Power, T.; Mulcahy, R. Supplementation with Carotenoids, Omega-3 Fatty Acids, and Vitamin E Has a Positive Effect on the Symptoms and Progression of Alzheimer’s Disease. J. Alzheimers Dis. 2022, 90, 233–249. [Google Scholar] [CrossRef]
  98. Yang, Q.N.; Deng, W.; Wu, D.T.; Li, J.; Liu, H.Y.; Yan, H.L.; Du, K.; Hu, Y.C.; Zou, L.; Huang, J.W. Characterization, Antioxidant Capacity, and Anti-Inflammatory Activity of Polyphenol-Enriched Extracts Obtained from Unripe, Mature, and Overripe Fruits of Red-Fleshed Kiwifruit Cultivars. Foods 2024, 13, 2860. [Google Scholar] [CrossRef]
  99. Tong, Q.; Yi, Z.; Ma, L.; Tan, Y.; Liu, D.; Cao, X.; Ma, X.; Li, X. Microenvironment-Responsive Antibacterial, Anti-Inflammatory, and Antioxidant Pickering Emulsion Stabilized by Curcumin-Loaded Tea Polyphenol Particles for Accelerating Infected Wound Healing. ACS Appl. Mater. Interfaces 2024, 16, 44467–44484. [Google Scholar] [CrossRef]
  100. Kurtz, J.A.; Feresin, R.G.; Grazer, J.; Otis, J.; Wilson, K.E.; Doyle, J.A.; Zwetsloot, K.A. Effects of Quercetin and Citrulline on Nitric Oxide Metabolites and Antioxidant Biomarkers in Trained Cyclists. Nutrients 2025, 17, 224. [Google Scholar] [CrossRef]
  101. Xia, W.; Chen, K.; Zhu, Y.Z.; Zhang, C.J.; Chen, Y.L.; Wang, F.; Xie, Y.Y.; Hider, R.C.; Zhou, T. Antioxidant and anti-tyrosinase activity of a novel stilbene analogue as an anti-browning agent. J. Sci. Food Agric. 2022, 102, 3817–3825. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, Y.; Zhang, X.; Yao, Y.; Huang, X.; Li, C.; Deng, P.; Jiang, G.; Dai, Q. The effect of epigallocatechin gallate on laying performance, egg quality, immune status, antioxidant capacity, and hepatic metabolome of laying ducks reared in high temperature condition. Vet. Q. 2023, 43, 1–11. [Google Scholar] [CrossRef] [PubMed]
  103. Yu, C.; Wang, D.; Yang, Z.; Wang, T. Pharmacological Effects of Polyphenol Phytochemicals on the Intestinal Inflammation via Targeting TLR4/NF-kappaB Signaling Pathway. Int. J. Mol. Sci. 2022, 23, 6939. [Google Scholar] [CrossRef]
  104. Kuriya, K.; Itoh, S.; Isoda, A.; Tanaka, S.; Nishio, M.; Umekawa, H. Green tea polyphenol EGCg induces cell fusion via reactive oxygen species. Biochem. Biophys. Rep. 2023, 35, 101536. [Google Scholar] [CrossRef]
  105. Ellis, L.Z.; Liu, W.; Luo, Y.; Okamoto, M.; Qu, D.; Dunn, J.H.; Fujita, M. Green tea polyphenol epigallocatechin-3-gallate suppresses melanoma growth by inhibiting inflammasome and IL-1beta secretion. Biochem. Biophys. Res. Commun. 2011, 414, 551–556. [Google Scholar] [CrossRef]
  106. Elmadhun, N.Y.; Sabe, A.A.; Robich, M.P.; Chu, L.M.; Lassaletta, A.D.; Sellke, F.W. The pig as a valuable model for testing the effect of resveratrol to prevent cardiovascular disease. Ann. N. Y Acad. Sci. 2013, 1290, 130–135. [Google Scholar] [CrossRef]
  107. Lyu, Y.L.; Zhou, H.F.; Yang, J.; Wang, F.X.; Sun, F.; Li, J.Y. Biological Activities Underlying the Therapeutic Effect of Quercetin on Inflammatory Bowel Disease. Mediat. Inflamm. 2022, 2022, 5665778. [Google Scholar] [CrossRef]
  108. Ojo, A.B.; Adanlawo, I.G. Antioxidant, antidiabetic, and anti-inflammatory activities of flavonoid-rich fractions of Solanum anguivi Lam. fruit: In vitro and ex vivo studies. Heliyon 2024, 10, e31895. [Google Scholar] [CrossRef]
  109. She, R.; Zhang, Z.; Han, M.; Zhao, D.; Li, X.; Zhou, J.; Chang, Y.; Zhang, X.; Li, X. Luteolin Exhibits Anxiolytic and Antidepressant Potential in Parkinson’s Disease Rat: Antioxidant and Anti-Inflammatory Effects. Rejuvenation Res. 2025, 28, 67–82. [Google Scholar] [CrossRef]
  110. Kuru Bektasoglu, P.; Demir, D.; Koyuncuoglu, T.; Yuksel, M.; Peker Eyuboglu, I.; Karagoz Koroglu, A.; Akakin, D.; Yildirim, A.; Celikoglu, E.; Gurer, B. Possible anti-inflammatory, antioxidant, and neuroprotective effects of apigenin in the setting of mild traumatic brain injury: An investigation. Immunopharmacol. Immunotoxicol. 2023, 45, 185–196. [Google Scholar] [CrossRef]
  111. Khalil, H.E.; Ibrahim, H.M.; Ahmed, E.A.; Emeka, P.M.; Alhaider, I.A. Orientin, a Bio-Flavonoid from Trigonella hamosa L., Regulates COX-2/PGE-2 in A549 Cell Lines via miR-26b and miR-146a. Pharmaceuticals 2022, 15, 154. [Google Scholar] [CrossRef] [PubMed]
  112. Monmai, C.; Kim, J.S.; Chin, J.H.; Lee, S.; Baek, S.H. Inhibitory Effects of Polyphenol- and Flavonoid-Enriched Rice Seed Extract on Melanogenesis in Melan-a Cells via MAPK Signaling-Mediated MITF Downregulation. Int. J. Mol. Sci. 2023, 24, 11841. [Google Scholar] [CrossRef]
  113. Ren, Q.; Guo, F.; Tao, S.; Huang, R.; Ma, L.; Fu, P. Flavonoid fisetin alleviates kidney inflammation and apoptosis via inhibiting Src-mediated NF-kappaB p65 and MAPK signaling pathways in septic AKI mice. Biomed. Pharmacother. 2020, 122, 109772. [Google Scholar] [CrossRef]
  114. Che, D.N.; Cho, B.O.; Kim, J.S.; Shin, J.Y.; Kang, H.J.; Jang, S.I. Luteolin and Apigenin Attenuate LPS-Induced Astrocyte Activation and Cytokine Production by Targeting MAPK, STAT3, and NF-kappaB Signaling Pathways. Inflammation 2020, 43, 1716–1728. [Google Scholar] [CrossRef]
  115. Kasahara, K.; Kerby, R.L.; Cross, T.L.; Everhart, J.; Kay, C.; Bolling, B.W.; Backhed, F.; Rey, F.E. Gut microbiota and diet matrix modulate the effects of the flavonoid quercetin on atherosclerosis. Res. Sq. 2023. [Google Scholar] [CrossRef]
  116. Chen, S.Y.; Zhou, Q.Y.; Chen, L.; Liao, X.; Li, R.; Xie, T. The Aurantii Fructus Immaturus flavonoid extract alleviates inflammation and modulate gut microbiota in DSS-induced colitis mice. Front. Nutr. 2022, 9, 1013899. [Google Scholar] [CrossRef] [PubMed]
  117. Tratensek, A.; Locatelli, I.; Grabnar, I.; Drobne, D.; Vovk, T. Oxidative stress-related biomarkers as promising indicators of inflammatory bowel disease activity: A systematic review and meta-analysis. Redox Biol. 2024, 77, 103380. [Google Scholar] [CrossRef]
  118. Inoue, H.; Shimamoto, S.; Takahashi, H.; Kawashima, Y.; Wataru, S.; Ijiri, D.; Ohtsuka, A. Effects of astaxanthin-rich dried cell powder from Paracoccus carotinifaciens on carotenoid composition and lipid peroxidation in skeletal muscle of broiler chickens under thermo-neutral or realistic high temperature conditions. Anim. Sci. J. 2019, 90, 229–236. [Google Scholar] [CrossRef] [PubMed]
  119. Berretta, M.; Quagliariello, V.; Bignucolo, A.; Facchini, S.; Maurea, N.; Di Francia, R.; Fiorica, F.; Sharifi, S.; Bressan, S.; Richter, S.N.; et al. The Multiple Effects of Vitamin D against Chronic Diseases: From Reduction of Lipid Peroxidation to Updated Evidence from Clinical Studies. Antioxidants 2022, 11, 1090. [Google Scholar] [CrossRef]
  120. Lee, Y.R.; Lee, W.H.; Lee, S.Y.; Lee, J.; Kim, M.S.; Moon, M.; Park, G.W.; Kim, H.S.; Kim, J.I.; Lee, J.S.; et al. Regulation of Reactive Oxygen Species Promotes Growth and Carotenoid Production Under Autotrophic Conditions in Rhodobacter sphaeroides. Front. Microbiol. 2022, 13, 847757. [Google Scholar] [CrossRef]
  121. Crawford, A.C.; Francki, M.G. Chromosomal location of wheat genes of the carotenoid biosynthetic pathway and evidence for a catalase gene on chromosome 7A functionally associated with flour b* colour variation. Mol. Genet. Genom. 2013, 288, 483–493. [Google Scholar] [CrossRef]
  122. Wu, H.; Li, S.; Wang, L.; Liang, J.; Yan, L.; Dong, J. Fucoxanthin, a Marine Carotenoid, Suppresses Mycoplasma pneumoniae-Triggered Inflammatory Cytokine Production and Promotes Bacterial Clearance in a Murine Model. Evid. Based Complement. Altern. Med. 2022, 2022, 6238162. [Google Scholar] [CrossRef] [PubMed]
  123. Meurer, M.C.; Mees, M.; Mariano, L.N.B.; Boeing, T.; Somensi, L.B.; Mariott, M.; da Silva, R.; Dos Santos, A.C.; Longo, B.; Santos Franca, T.C.; et al. Hydroalcoholic extract of Tagetes erecta L. flowers, rich in the carotenoid lutein, attenuates inflammatory cytokine secretion and improves the oxidative stress in an animal model of ulcerative colitis. Nutr. Res. 2019, 66, 95–106. [Google Scholar] [CrossRef] [PubMed]
  124. Baz, L.; Algarni, S.; Al-Thepyani, M.; Aldairi, A.; Gashlan, H. Lycopene Improves Metabolic Disorders and Liver Injury Induced by a Hight-Fat Diet in Obese Rats. Molecules 2022, 27, 7736. [Google Scholar] [CrossRef]
  125. Yang, X.; Li, L.; Shang, G.; Zhuo, M.; Zhu, H.; Xu, S.; Zhao, J.; Hou, X.; Shi, Y. Oral Curcumin through Mesoporous Silica Nanomaterials with Distinct Morphologies: Synthesis, Characterization, Biosafety Evaluation, and Antioxidant Activity In Vivo. Langmuir 2024, 40, 27216–27227. [Google Scholar] [CrossRef]
  126. Zhang, X.; Zhang, H.; Wang, J.; Chen, Y.; Lin, J.; Wang, Q.; Wu, C.; Chen, H.; Lin, Y. Curcumin attenuates ulcerative colitis via regulation of Sphingosine kinases 1/NF-kappaB signaling pathway. Biofactors 2025, 51, e70001. [Google Scholar] [CrossRef] [PubMed]
  127. Sarkar, E.; Kotiya, A.; Bhuyan, R.; Raza, S.T.; Misra, A.; Ahmad, R.; Mahdi, A.A. Curcumin chemo-sensitizes intrinsic apoptosis through ROS-mediated mitochondrial hyperpolarization and DNA damage in breast cancer cells. Cell Signal 2025, 128, 111637. [Google Scholar] [CrossRef]
  128. Wang, Y.; Guo, R.; Zou, M.; Jiang, L.; Kong, L.; Zhao, S.; Zhang, X.; Wang, W.; Xu, B. Combined ROS Sensitive Folate Receptor Targeted Micellar Formulations of Curcumin Effective Against Rheumatoid Arthritis in Rat Model. Int. J. Nanomed. 2024, 19, 4217–4234. [Google Scholar] [CrossRef]
  129. Alsahli, M.A.; Almatroodi, S.A.; Almatroudi, A.; Khan, A.A.; Anwar, S.; Almutary, A.G.; Alrumaihi, F.; Rahmani, A.H. 6-Gingerol, a Major Ingredient of Ginger Attenuates Diethylnitrosamine-Induced Liver Injury in Rats through the Modulation of Oxidative Stress and Anti-Inflammatory Activity. Mediat. Inflamm. 2021, 2021, 6661937. [Google Scholar] [CrossRef]
  130. Isa, Y.; Miyakawa, Y.; Yanagisawa, M.; Goto, T.; Kang, M.S.; Kawada, T.; Morimitsu, Y.; Kubota, K.; Tsuda, T. 6-Shogaol and 6-gingerol, the pungent of ginger, inhibit TNF-alpha mediated downregulation of adiponectin expression via different mechanisms in 3T3-L1 adipocytes. Biochem. Biophys. Res. Commun. 2008, 373, 429–434. [Google Scholar] [CrossRef]
  131. Lee, T.Y.; Lee, K.C.; Chen, S.Y.; Chang, H.H. 6-Gingerol inhibits ROS and iNOS through the suppression of PKC-alpha and NF-kappaB pathways in lipopolysaccharide-stimulated mouse macrophages. Biochem. Biophys. Res. Commun. 2009, 382, 134–139. [Google Scholar] [CrossRef] [PubMed]
  132. Siddique, R.; Muhammad, F.; Faisal, M.N.; Akhtar, B.; Saleem, A.; Kousar, S.; Sharif, A.; Saeed, M.; Muhammad, S. Gingerol nanoparticles attenuate complete Freund adjuvant-induced arthritis in rats via targeting the RANKL/OPG signaling pathway. Inflammopharmacology 2024, 32, 3311–3326. [Google Scholar] [CrossRef]
  133. Sahler, J.; Bernard, J.J.; Spinelli, S.L.; Blumberg, N.; Phipps, R.P. The Feverfew plant-derived compound, parthenolide enhances platelet production and attenuates platelet activation through NF-kappaB inhibition. Thromb. Res. 2011, 127, 426–434. [Google Scholar] [CrossRef] [PubMed]
  134. Najera-Martinez, M.; Lara-Vega, I.; Avilez-Alvarado, J.; Pagadala, N.S.; Dzul-Caamal, R.; Dominguez-Lopez, M.L.; Tuszynski, J.; Vega-Lopez, A. The Generation of ROS by Exposure to Trihalomethanes Promotes the IkappaBalpha/NF-kappaB/p65 Complex Dissociation in Human Lung Fibroblast. Biomedicines 2024, 12, 2399. [Google Scholar] [CrossRef] [PubMed]
  135. Shang, Y.; Han, D.; Deng, K.; Zhou, H.; Wu, M. Quercetin Boosts Pulsatile Gonadotropin-Releasing Hormone Release to Improve Luteal Function via Inhibiting NF-kappaB/NLRP3-Mediated Neuron Pyroptosis. Mol. Nutr. Food Res. 2024, 68, e2400649. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, D.; Wang, W.; Ou, H.; Ning, J.; Zhou, Y.; Ke, J.; Hou, A.; Chen, L.; Li, P.; Ma, Y.; et al. Identification of chalcone analogues as anti-inflammatory agents through the regulation of NF-kappaB and JNK activation. RSC Med. Chem. 2024, 15, 2002–2017. [Google Scholar] [CrossRef]
  137. Ma, Y.; Ma, Z.; Zhang, Y.; Luo, C.; Huang, P.; Tong, J.; Ding, H.; Liu, H. Apigenin and baicalein ameliorate thoracic aortic structural deterioration and cognitive deficit via inhibiting AGEs/RAGE/NF-kappaB pathway in D-galactose-induced aging rats. Eur. J. Pharmacol. 2024, 976, 176660. [Google Scholar] [CrossRef]
  138. Hossen, I.; Kaiqi, Z.; Hua, W.; Junsong, X.; Mingquan, H.; Yanping, C. Epigallocatechin gallate (EGCG) inhibits lipopolysaccharide-induced inflammation in RAW 264.7 macrophage cells via modulating nuclear factor kappa-light-chain enhancer of activated B cells (NF-kappaB) signaling pathway. Food Sci. Nutr. 2023, 11, 4634–4650. [Google Scholar] [CrossRef]
  139. Liu, Y.; Liu, B.; Shi, M.; Ye, T.; Li, H. NLRP3 Inflammasome Activation Is Involved in Geniposide-Induced Hepatotoxicity. Mediat. Inflamm. 2025, 2025, 4112856. [Google Scholar] [CrossRef]
  140. Zu, R.; Lu, H.; Liu, W.; Shao, S.; Zheng, J.; Ying, X.; Zhou, Y.; Li, Z.; Wang, W.; Li, D.; et al. Research Progress in the Molecular Mechanism of NLRP3 Inflammasome in Alzheimer’s Disease and Regulation by Natural Plant Products. Mol. Neurobiol. 2025, 62, 7296–7312. [Google Scholar] [CrossRef]
  141. Yang, K.; Zeng, L.; He, Q.; Wang, S.; Xu, H.; Ge, J. Advancements in research on the immune-inflammatory mechanisms mediated by NLRP3 inflammasome in ischemic stroke and the regulatory role of natural plant products. Front. Pharmacol. 2024, 15, 1250918. [Google Scholar] [CrossRef] [PubMed]
  142. Santos-Garcia, I.; Bascunana, P.; Brackhan, M.; Villa, M.; Eiriz, I.; Bruning, T.; Pahnke, J. The ABC transporter A7 modulates neuroinflammation via NLRP3 inflammasome in Alzheimer’s disease mice. Alzheimers Res. Ther. 2025, 17, 30. [Google Scholar] [CrossRef]
  143. Sharma, P.; Chouhan, R.; Bakshi, P.; Gandhi, S.G.; Kaur, R.; Sharma, A.; Bhardwaj, R. Amelioration of Chromium-Induced Oxidative Stress by Combined Treatment of Selected Plant-Growth-Promoting Rhizobacteria and Earthworms via Modulating the Expression of Genes Related to Reactive Oxygen Species Metabolism in Brassica juncea. Front. Microbiol. 2022, 13, 802512. [Google Scholar] [CrossRef] [PubMed]
  144. Farhan, M. Cytotoxic Activity of the Red Grape Polyphenol Resveratrol against Human Prostate Cancer Cells: A Molecular Mechanism Mediated by Mobilization of Nuclear Copper and Generation of Reactive Oxygen Species. Life 2024, 14, 611. [Google Scholar] [CrossRef]
  145. Wang, Y.; Wang, J.; Jiao, Y.; Chen, K.; Chen, T.; Wu, X.; Jiang, X.; Bu, W.; Liu, C.; Qu, X. Redox-active polyphenol nanoparticles deprive endogenous glutathione of electrons for ROS generation and tumor chemodynamic therapy. Acta Biomater. 2023, 172, 423–440. [Google Scholar] [CrossRef] [PubMed]
  146. Ahuja, K.D.; Pittaway, J.K.; Ball, M.J. Effects of olive oil and tomato lycopene combination on serum lycopene, lipid profile, and lipid oxidation. Nutrition 2006, 22, 259–265. [Google Scholar] [CrossRef]
  147. Andrade, P.; Santamarina, A.B.; de Freitas, J.A.; Marum, A.; Pessoa, A.F.M. Personalized nutrition and precision medicine in perimenopausal women: A minireview of genetic polymorphisms COMT, FUT2, and MTHFR. Clinics 2025, 80, 100549. [Google Scholar] [CrossRef]
  148. Gardner, D.S.L.; Saboo, B.; Kesavadev, J.; Mustafa, N.; Villa, M.; Mahoney, E.; Bajpai, S. Digital Health Technology in Diabetes Management in the Asia-Pacific Region: A Narrative Review of the Current Scenario and Future Outlook. Diabetes Ther. 2025, 16, 349–369. [Google Scholar] [CrossRef]
  149. Giri, N.A.; Gaikwad, P.; Gaikwad, N.N.; Manjunatha, N.; Krishnakumar, T.; Kad, V.; Raigond, P.; Suryavanshi, S.; Marathe, R.A. Development of fiber-enriched muffins using pomegranate peel powder and its effect on physico-chemical properties and shelf life of the muffins. J. Sci. Food Agric. 2024, 104, 2346–2358. [Google Scholar] [CrossRef]
  150. Ben-Othman, S.; Joudu, I.; Bhat, R. Bioactives From Agri-Food Wastes: Present Insights and Future Challenges. Molecules 2020, 25, 510. [Google Scholar] [CrossRef]
  151. Nunez-Gomez, V.; Gonzalez-Barrio, R.; Baenas, N.; Moreno, D.A.; Periago, M.J. Dietary-Fibre-Rich Fractions Isolated from Broccoli Stalks as a Potential Functional Ingredient with Phenolic Compounds and Glucosinolates. Int. J. Mol. Sci. 2022, 23, 13309. [Google Scholar] [CrossRef] [PubMed]
  152. Slavin, J.L. Dietary fiber and body weight. Nutrition 2005, 21, 411–418. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Plant-derived bioactive compounds improve our health.
Figure 1. Plant-derived bioactive compounds improve our health.
Ijms 26 05206 g001
Figure 2. The main plant-derived bioactive compounds discussed in this paper.
Figure 2. The main plant-derived bioactive compounds discussed in this paper.
Ijms 26 05206 g002
Table 1. Plant-derived bioactive compounds and their rich plant sources.
Table 1. Plant-derived bioactive compounds and their rich plant sources.
High Content PlantsPrincipal Effect
QuercetinApples, onionAntioxidant, anti-inflammatory effect
Epigallocatechin gallateGreen tea leavesAntioxidant, anti-inflammatory effect
LuteolinCelery, bell peppersAntioxidant, anti-inflammatory effect
β-caroteneCarrots, squashAntioxidant, anti-inflammatory effect
LycopeneTomatoes, watermelonAnti-inflammatory, and reduction the risk of obesity-related diseases effect
LuteinSpinach, kaleAnti-inflammatory effect
CurcuminCurcuma longaAntioxidant, anti-inflammatory effect
GingerolZingiber officinaleAnti-inflammatory effect
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

Nakadate, K.; Ito, N.; Kawakami, K.; Yamazaki, N. Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications. Int. J. Mol. Sci. 2025, 26, 5206. https://doi.org/10.3390/ijms26115206

AMA Style

Nakadate K, Ito N, Kawakami K, Yamazaki N. Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications. International Journal of Molecular Sciences. 2025; 26(11):5206. https://doi.org/10.3390/ijms26115206

Chicago/Turabian Style

Nakadate, Kazuhiko, Nozomi Ito, Kiyoharu Kawakami, and Noriko Yamazaki. 2025. "Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications" International Journal of Molecular Sciences 26, no. 11: 5206. https://doi.org/10.3390/ijms26115206

APA Style

Nakadate, K., Ito, N., Kawakami, K., & Yamazaki, N. (2025). Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications. International Journal of Molecular Sciences, 26(11), 5206. https://doi.org/10.3390/ijms26115206

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop