Next Article in Journal
Answer to Controversy: miR-10a Replacement Approaches Do Not Offer Protection against Chemotherapy-Induced Gonadotoxicity in Mouse Model
Next Article in Special Issue
Lysozyme-Induced Transcriptional Regulation of TNF-α Pathway Genes in Cells of the Monocyte Lineage
Previous Article in Journal
OsATG8c-Mediated Increased Autophagy Regulates the Yield and Nitrogen Use Efficiency in Rice
Previous Article in Special Issue
Effects of Extract of Arrabidaea chica Verlot on an Experimental Model of Osteoarthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Inflammation by Flavonoids: Novel Therapeutic Strategy for Metabolic Disorders

1
Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
2
Department of Biochemistry, University of Allahabad, Allahabad 211 002, India
3
Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
4
Bioorganic Chemistry Laboratory, Facultad de Ciencias Básicas y Aplicadas, Campus Nueva Granada, Universidad Militar Nueva Granada, Cajicá 250247, Colombia
5
Research Institute of Biotechnology and Medical Converged Science, Dongguk University-Seoul, Ilsandong-gu, Gyeonggi-do 10326, Korea
6
Post Graduate Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar 751 004, Odisha, India
7
Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy
8
Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
9
Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy
10
Medicinal Plants Research Center, Institute of Medicinal Plants, Karaj 141554364, Iran
11
Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran 141556451, Iran
12
Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(19), 4957; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20194957
Submission received: 29 August 2019 / Revised: 28 September 2019 / Accepted: 30 September 2019 / Published: 8 October 2019
(This article belongs to the Special Issue Natural Anti-inflammatory Agents 2019)

Abstract

:
A balanced metabolic profile is essential for normal human physiological activities. Disproportions in nutrition give rise to imbalances in metabolism that are associated with aberrant immune function and an elevated risk for inflammatory-associated disorders. Inflammation is a complex process, and numerous mediators affect inflammation-mediated disorders. The available clinical modalities do not effectively address the underlying diseases but rather relieve the symptoms. Therefore, novel targeted agents have the potential to normalize the metabolic system and, thus, provide meaningful therapy to the underlying disorder. In this connection, polyphenols, the well-known and extensively studied phytochemical moieties, were evaluated for their effective role in the restoration of metabolism via various mechanistic signaling pathways. The various flavonoids that we observed in this comprehensive review interfere with the metabolic events that induce inflammation. The mechanisms via which the polyphenols, in particular flavonoids, act provide a promising treatment option for inflammatory disorders. However, detailed clinical studies of such molecules are required to decide their clinical fate.

1. Introduction

Multicellular organisms fight infections, manage different external and internal damages, and maintain the body’s energy balance, particularly under energy deficit conditions. In overall summation, the immune system and metabolic pathways are among the primary essentialities, without which the animal kingdom would cease to exist [1]. Furthermore, both the immune and metabolic pathways co-evolved in a manner in which they are closely linked and interdependent. Nutritional imbalances disrupt metabolism, leading to irregular immune function and an elevated risk for inflammatory-associated disorders [2]. In ancient history, inflammation was characterized based on the visual observations of five cardinal signs specifically named as rubor (redness), tumor (swelling), calor (heat), dolor (pain), and lastly function laesa (loss of function) [3]. In short, inflammation could be described as a response of living tissue to local injury [4,5]. There is evidence to support that inflammation plays a decisive role in neoplastic progression. This concept is based on the relationship between incessant inflammatory activities due to viruses, bacteria, parasites, infections, and carcinogenesis and their effects within the organs and tissues [6,7,8,9,10].
Inflammation can be further divided into acute and chronic. The local effects of acute inflammation include modifications of metabolic and functional activities of polymorphonuclear cells and macrophages. The systemic effects of acute inflammation include modification of immune response and non-specific defenses against infection and neoplasia [11]. Acute inflammation can also lead to fever and leukocytosis [4]. Chronic inflammation is characterized by the medical condition of chronic inflammatory disease. This medical condition can be defined as a lengthy and persistent pro-inflammatory state marked specifically by the formation of new connective tissue [12]. There are many diseases included in this category: autoimmune disease, metabolic syndromes, neurodegenerative disease, chronic inflammatory bowel disease, chronic obstructive pulmonary disease, and cardiovascular disease (CVD) [12,13]. Among the abovementioned diseases and syndromes, metabolic syndromes are strongly associated with chronic inflammation [1]. Inflammatory reaction pathways involve various receptors and molecules, such as Toll-like receptors (TLRs) or nucleotide oligomerization domain (NOD)-like receptors (NLRs). These receptors activate major mitogen-activated protein kinase (MAPK) cascades and stimulate translocation of regulatory nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) [7]. It was documented that the expression of pro-inflammatory cytokine mediators orchestrates an indispensable role in various CVDs, metabolic syndromes, and atherosclerosis [14,15,16,17,18].
A number of investigators reported the natural amelioration of inflammation via the use of polyphenols due to their anti-inflammatory activities [19,20,21]. It was found that numerous polyphenols, such as flavonoids, are active suppressors of inflammatory cytokines, modulators of transcription factors and inflammation-related pathways, and reducers of accumulated of nitric oxide (NO) or reactive oxygen species (ROS) [22]. For example, a species-specific flavonoid, glabridin, was found to attenuate mediators of inflammation including nitric oxide (NO), tumor necrosis factor-α (TNF-α), and interlukin (IL)-1β in THP-1 cells, RAW 264.7 cells, and J774a.1 cells [23,24,25,26]. Another investigator showed that glabridin also inhibited the maturation of dendritic cells by blocking NF-κB and MAPK signaling cascade [27]. Similarly, phloretin, another important polyphenol, was found to inhibit expression of IL-8, C–X–C motif chemokine 10, and TNF-α messenger RNAs (mRNAs) [28].
Oxidative stress and inflammatory mediators are well known for their role in generating ROS and reactive nitrogen species (RNS) by affecting NADPH oxidase and nitric oxide synthase (NOS) respectively. ROS trigger redox-sensitive kinases, such as apoptosis signal-regulating kinase 1 (ASK1), which in turn activates downstream MAPKs, NF-κB, and activator protein 1 (AP-1), resulting in the induction of inflammatory gene expression. Research shows that phenolic phytochemicals possess strong antioxidant activity due to the presence of hydroxyl groups within their aromatic rings [29,30]. The molecular mechanism involved lies in the capacity of the phenolic phytochemicals to increase the level of anti-inflammatory genes, such as glutathione peroxidase (GPx), superoxide dismutase (SOD), and hemeoxygenase-1 (HO-1), via activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) [30].
To manage metabolic syndromes, global strategies initially focused on lifestyle changes such as diet and physical activity. Diets rich in plant-derived products with a high content of bioactive compounds, mono/polyunsaturated fatty acids, and polyphenols were shown to lower related risks of metabolic syndrome [31,32,33]. Polyphenols are available in various regular diets, indicating negligible side effects if taken in a controlled manner. This fact makes them a good candidate for their use in various clinical trials in treatment of various diseases. There is a large body of evidence to suggest that polyphenols play an effective role as anti-inflammatory agents in various metabolic diseases [34,35,36]. However, there is no review article that specifically concentrates on this subject. In addition, there is little information on the utilization of polyphenols to ameliorate the effect of metabolic disorders, as well as their in vitro and in vivo evaluation.
The purpose of this review is to summarize recent data on dietary polyphenols, with a special emphasis on flavonoids, which affect the inflammation involved in various metabolic disorders, and to gather the results of clinical studies on the chronic supplementation of flavonoids on metabolic syndromes features.

2. Inflammation and Metabolic Disorders

Inflammation is a defensive strategy developed in higher organisms in reaction to the harmful effects of tissue injury, microbial infection, and other detrimental conditions. It is an important immune response by the host that aids in tissue repair and the elimination of harmful stimuli [37]. However, long-term inflammation is often harmful and can cause several metabolic diseases [13] (Figure 1).
Moreover, high caloric intake coupled with inactive lifestyle leads to increased incidence of obesity, type 2 diabetes, and cardiovascular diseases, which are associated with chronic diseases [38,39,40]. Metabolism and inflammation are interrelated. Metabolic disorders display a strong inflammatory foundation, and inflammation is also linked to metabolic changes. The complex interaction between inflammatory and metabolic pathways is underscored by the biological and functional similarity of macrophages and adipocytes. In both cell types, similar gene expression is observed. Several adipocyte proteins are expressed by macrophages such as peroxisome proliferator-activated receptor-γ (PPARγ), adiponectin protein 2 (aP2), and adipocyte/macrophage fatty-acid binding protein (FABP). In addition, adipocytes express many “macrophage” gene products, such as IL-6, TNF-α, and matrix metalloproteinases (MMPs) [41,42].
Monocytes at infection sites differentiate into macrophages under the influence of the local cytokine environment [43]. Based on function and cytokine expression, differentiated macrophages are classified as M1 and M2 macrophages. M1 macrophages produce pro-inflammatory cytokines such as IL-1, IL-6, and TNF-α, which result in the mobilization of neutrophils and help to initiate the innate immune response against pathogens. Furthermore, T cells are stimulated by the cumulative action of pro-inflammatory cytokines and their ligands. M1 macrophages and T lymphocytes are linked to chronic inflammation in metabolic disorders. M2 macrophages are anti-inflammatory mediators that participate in wound healing and the angiogenic process [44].
The transcription factor NF-κB is a well-known inflammatory and immunological mediator that connects inflammation to metabolic responses. It helps to better understand metabolic diseases and provides insight into the development of therapeutic strategies. Pathogen-associated molecular patterns (PAMPs) and cytokines trigger cell surface receptors, including TLRs, which initiate a signaling pathway that activates NF-κB [45]. NF-κB-regulated gene expression is responsible for the differentiation of various types of immune cells. Involvement of NF-κB was revealed in three common metabolic disorders: atherosclerosis, obesity, and insulin resistance. Extracellular stimuli control the activity of NF-κB. The nuclear localization and transcriptional function of NF-κB is prevented by inhibitor of κB (IκB). IkB sequesters NF-κB in the cytoplasm of resting cells. Interaction of cytokine or PAMP with cell surface receptors begins a signaling sequence that activates the inhibitor of κB kinase (IKK) complex. Phosphorylation of IκB by IKK stimulates its break down that releases NF-κB and, consequently, NF-κB moves inside the nucleus and promotes transcription of target genes. Research findings suggest that the IKKγ/IKKβ complex initiates NF-κB-regulated gene expression downstream of TLRs and cytokine receptors [45].
Most resting cells do not express the IKK3, but NF-κB induces its transcription downstream of inflammatory stimuli [46]. Research findings showed the involvement of the non-canonical IKK kinase IKK3 in high-fat diet-induced obesity. In response to excess nutrients, IKK3 expression increases approximately 40 times in fat cells and fat-infiltrating macrophages [47].
Deficiency of IKK3 causes uncoupling of obesity from a fat-rich diet by increasing energy consumption, thermogenesis, and aerobic respiration. IKK3 knockdown mice are protected from pro-inflammatory pathway activation, chronic inflammation in liver and adipose tissue, and diet-induced insulin resistance because of lesser weight gain and possibly improper cytokine production and signaling pathway [48,49,50,51,52]. Hence, IKK family signaling cascade performs a vital function in overnutrition-induced obesity, as well as in metabolic disease.
Inflammatory processes are recognized to induce atherosclerosis. Atherosclerosis begins with the accumulation of excess unmetabolized lipoproteins in the blood. Oxidized lipoproteins induce vascular endothelia to release chemokines, macrophage inflammatory protein-1α and monocyte chemoattractant protein-1 (MCP-1), which attract leukocytes to the inflammation site [48,49]. Association of oxidative stress and mitochondrial dysfunction with inflammation is implicated in type 2 diabetes, leading to insulin resistance in muscle and adipocyte cells, as well as impaired insulin secretion by the β cells of the islets of Langerhans [50,51]. Oxidative stress activates NF-κB and c-Jun N-terminal kinase (JNK) pathways and, thus, shows the ability of ROS to cause insulin resistance [52,53,54].

2.1. Inflammation in Cardiovascular Disease

Heart disease and stroke are responsible for many of deaths yearly. Hypertension, dyslipidemia, diabetes mellitus (DM), visceral obesity, and smoking are cardiovascular risk factors which contribute to cardiovascular disease [55]. Lipid-lowering drugs in combination with blood pressure-lowering pharmacological agents and anti-thrombotic drugs can reduce the risk of atherosclerotic vascular disease. In spite of these therapeutic advances, fresh approaches are still required to manage the increasing incidence of atherosclerosis. Immunity (innate and acquired) plays an essential role in the beginning, advancement, and instability of the atherosclerotic plaque development. However, for the prevention of CVD, it is very difficult to target a single pathway due to the considerable complexity of the immune system network [56].
Inflammation is related to the development of several cardiovascular complications. Increased levels of inflammatory markers, such as C-reactive protein (CRP) and serum amyloid A (SAA), are suggested as predictive markers of future cardiovascular disease development [57]. A drawback related to the currently used biomarkers of inflammation in CVD is their non-specificity (e.g., CRP). Therefore, a major challenge for researchers is to identify specific markers of inflammation that are relevant to the pathophysiology of disease and that can be used to guide targeted therapies [57].

2.1.1. Acute Myocardial Infraction (AMI)

Coronary atherosclerotic plaque rupture leads to the speedy formation of a thrombus in the infarct-associated epicardial artery and is coupled with the inhibition of blood supply distal to the obstruction site, resulting in an AMI. Plaque erosions and calcific nodules were also suggested as other causes of AMI [58]. An organized immune response begins as a result of myocardial ischemia and occurs locally at the injury site, as well as in the systemic circulation and at distant sites [57]. The immune reactions serve dual roles as mediators of injury and, afterward, as facilitators of repair and recovery. Continuous rising levels of nonspecific inflammatory mediators, e.g., IL-6 and high-sensitivity CRP in plasma, are observed in severe cases [59,60].

Inflammatory Pathway in AMI

Myocardial ischemia causes injury to cardiomyocytes and, if ischemia persists for a long duration, death ensues with the liberation of intracellular contents, together with alterations in the extracellular matrix. Subsequently, endogenous signals initiate a cytokine “burst”, followed by platelet activation-mediated leukocyte stimulation and tissue permeation. This further leads to the quick mobilization of neutrophils to the site of myocardial injury followed by their degranulation, which causes direct damage to endothelial cells along with production of cytokines, ROS, and proteolytic enzymes [61]. As a result, the expression of leukocyte and platelet adhesion molecules elevates in endothelial cells, which further accelerates the binding and transmigration of monocytes in circulation [62]. This process overlaps with the reduction of in situ neutrophils because of local cell death coupled with egression of cells [63].
Numerous endogenous signals, namely, low-molecular hyaluronic acid, heat-shock proteins, fibronectin fragments, and high-mobility group protein B1, are frequently denoted as “danger-associated molecular patterns” (DAMPs). These DAMPS can trigger innate immune pathways, comprising the complement cascade, TLRs, ROS production, and nucleotide-binding domain leucine-rich repeat-containing receptors (NLRs). The specific TLR expression in cardiomyocytes (TLR4), an important characteristic feature of activated macrophages, is considerably elevated during interaction with DAMPs [57].
Animal experiments showed that reduction in infarct size and systemic inflammation, as well as improved left ventricular remodeling, is associated with TLR4 deficiency [64,65]. Subsequent to AMI, activation of TLR4 in patient monocytes suggests progression toward heart failure. On the other hand, TLR2 deficiency is related to decreased myocardial fibrosis after AMI and enhanced left ventricular remodeling. It appears that TLR4 plays a vital role in the initial inflammatory response, while TLR2 is crucial to left ventricular remodeling and repair [66]. Amongst various NLRs, the nucleotide oligomerization domain (NOD or NACHT), leucine-rich repeats (LRR) and pyrin domain (PYD) domains-containing protein 3 (NLRP3)–inflammasome complex is recognized as a key facilitator of injury after myocardial ischemia. In addition to NLRP3, the complex includes the cysteine protease caspase-1 and the apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD). The activated NLRP3–inflammasome binds and stimulates caspase-1, which transforms IL-1β to an active state, and is a key factor in the activation of various cytokines [67].
The complement cascade plays a major role in steering neutrophil and monocyte accumulation deeply into the damaged myocardium via the downstream complement effectors C3a and C5a. In addition, it also helps macrophages, microvascular endothelial cells, and vascular smooth muscle cells (VSMCs) in releasing monocyte chemotactic protein 1, which is also known as C–C motif chemokine 2 (CCL2) [68]. After AMI, ischemic cardiomyocytes release ROS immediately, which induce elevated levels of leukocyte chemokine, leading to an augmented capability of endothelial intercellular adhesion molecule1 (ICAM1) ligands to bind to neutrophils, activation of complement, and the accumulation of extra subpopulations of leukocytes that consequently become activated. Many cytokines, such as TNF-α, IL-1β, and IL-6, also act as key molecules in contributing to the pathophysiology of AMI [69].

2.1.2. Atherosclerosis

Atherosclerosis is an inflammation-participating disease in all of its stages, that is, starting from initial lesions until the end-stage thrombotic complications [70]. It results from complex phenomena comprising the interaction of lipoproteins, blood cells, arterial wall components, and the immune system. The process of advanced atherogenic plaque development from the earliest foam cell formation involves interplay of endothelial cells, smooth muscle cells (SMCs), lymphocytes, monocytes, and macrophages. This interaction is regulated by several inflammatory mediators associated with chronic low-grade inflammation [71]. The adhesion of leukocytes to the innermost surface of the artery wall is prevented by to the presence of endothelial cells (ECs) [72]. The role of these inflammatory molecules as proxy biomarkers and causative agents in the atherogenic development and plaque vulnerability was studied by our groups. The intimate linkage between atherosclerosis and inflammation is supported by experimental evidence. Several randomized controlled trials revealed that routine use of statins for prevention of CVD mediates their effect through anti-inflammatory action [73].
It is evidenced that smoking, hypertension, high saturated-fat diet consumption, hyperglycemia, obesity, or insulin resistance could induce the expression of a range of cytokines including TNF-α, TNF-β, IL-1α, IL-1β, IL-6, and certain messenger cytokines, such as macrophage colony stimulating factor (M-CSE), MCP-1, IL-18, and vascular cell adhesion molecule-1 (VCAM-1), by ECs. Induction of these molecules leads to the activation and recruitment of monocytes during atherosclerosis [70,72]. VCAM-1 specifically binds to T lymphocytes and monocytes in an early stage of atherosclerotic plaque formation [74]. VCAM-1 expression is induced by oxidized lipids through a pathway mediated by NF-κB and certain pro-inflammatory cytokines, such as IL-1β and TNF-α [70,75]. Furthermore, through diapedesis, T lymphocytes and monocytes penetrate the endothelial lining and enter the intima of the vessel. A basic requirement of this process is a chemoattractant gradient, which involves C–C chemokine receptors (CCR) sensing MCP-1/CCL2, a member of the C–C chemokine family [70,76].
Inflammation-induced alterations in lipoprotein function increase the risk of atherosclerosis. There are reports that inflammation and infection have an association with low-density lipoprotein. However, the LDL levels do not steadily rise and may even decline with inflammation and infection [77].
Inflammation and infection are responsible for disturbing the anti-atherogenic activity of serum high-density lipoprotein (HDL), in addition to reducing the serum HDL level [77]. Moreover, inflammation and infection also unfavorably affect most of the steps in the reverse cholesterol transport pathway, which plays a vital role in averting cholesterol build-up in macrophages and, hence, reduces the atherosclerotic process [78]. The initial event under the influence of inflammatory cytokines starts with the decreased production of the key protein component of HDL, Apo A-1. Pro-inflammatory cytokines in macrophages further reduce the expression of apolipoprotein E, scavenger receptor class B type I, ATP-binding cassette transporter A1 (ABCA1), and ATP Binding cassette subfamily G member 1, which in turn causes a reduction in the phospholipid and cholesterol efflux from the macrophage to HDL. The transformed HDL molecules synthesized during inflammation are poor acceptors of cellular cholesterol, and they may actually perform the opposite action by delivering cholesterol to the macrophage [79,80].
SR-B1 also facilitates cholesterol uptake by hepatocytes from HDL particles. Its expression in the liver is decreased by pro-inflammatory cytokines. Paraoxonase, an HDL-associated enzyme, plays a major role in the prevention of LDL oxidation. Oxidized LDL is pro-atherogenic and is more easily taken up by macrophages. Inflammatory response decreases the level of paraoxonase 1 expression in the liver and brings about a decline in circulating paraoxonase activity [77].
Aberrant stress activates the endothelium. It results in enhanced permeability to lipoproteins and increased expression of adhesion receptors, which enables deposition of lipid in the sub-endothelial space from apolipoprotein B (apoB)-containing lipoproteins. Activation of the endothelium also stimulates the accumulation of circulating monocytes originating from the spleen or bone marrow [81].
Monocyte adhesion is governed by the elevated expression of numerous cell adhesion molecules on the luminal surface of the endothelium, such as VCAM1, P-selectin, and ICAM1. This is followed by the expression of facilitators for the transmigration process belonging to three main chemokine families, i.e., CCR2, CCR5, and CX3C chemokine receptor 1 (CX3CR1) [82]. After recruitment, monocytes can directly affect the phenotype of in situ cells, differentiate, and locally multiply into distinct functional phenotypes, or terminally differentiate into macrophages. Activated macrophages take up lipids present in apoB-containing lipoproteins via several processes comprising pinocytosis of LDL, phagocytosis of aggregated LDL, uptake of altered apoB-lipoproteins by scavenger receptors, and cluster of differentiation 36-mediated uptake [57].
Continuous lipid build-up prompts their conversion into macrophage-derived foam cells. With the growing population of foam cells within arterial wall lesions, the level of accumulation surpasses the rate of clearance and, ultimately, the foam cells merge into a lipid-rich necrotic core. Interplay of cells from both the arms of immunity, e.g., the macrophages (innate immunity) and T and B lymphocytes, as well as dendritic cells (adaptive immunity), control the local inflammatory environment with the involvement of numerous cytokines in this process [83].
Based on their local milieu, T lymphocytes can be triggered to release pro-inflammatory T helper type 1 (Th1) cytokines (IL-1, IL-6, and TNF-α) or Th2 cytokines (IL-4, IL-10, and IL-13). The p38 MAPK/NF-κB pathways are mainly involved in IL-1 and TNF signaling. On the contrary, signal transducing protein gp130 activates IL-6 signaling, which in turn triggers Janus kinase 1 and signal transducer and activator of transcription 1 (STAT1) and STAT3, ultimately resulting in the stimulation of macrophages and endothelial cells to produce adhesion molecules and chemokines [84]. Propagation of disease is also attributed to the features within the plaque. New vessel formation in human plaques (neo-vascularization), initiating from the vasa vasorum, is theorized to cause intraplaque hemorrhage. This not only hastens plaque extension and inflammation, but also envisages impending plaque break [85]. The hypoxic environment inside the lipid-rich necrotic core induces hypoxia-inducible factor (HIF) 1α, which in turn activates vascular endothelial growth factor, ultimately leading to angiogenesis in advanced atheroma [86].
In both mouse and human macrophages, HIF1α and hypoxia were shown to cause altered lipid handling and suppression of cholesterol efflux in vitro via ABCA1. Furthermore, hypoxia and HIF1α were also reported as the driving force for higher glucose uptake, increased metabolic activity, and macrophage polarization in human atheroma during in vitro and ex vivo experiments [87].
Generally, endothelial cells (ECs) resist adhesion by leukocytes. However, several factors such as consumption of a high saturated-fat diet, hyperglycemia, hypertension, insulin resistance, obesity, or smoking can trigger the expression of adhesion molecules by ECs, thereby permitting the attachment of leukocytes to the arterial wall.

2.2. Inflammation in Diabetes and Insulin Resistance

Diabetes mellitus (DM) is specifically characterized by a moderate or absolute lack of insulin, leading to hyperglycemia [1]. DM can be broadly categorized into type 1 and type 2 diabetes. The former is an autoimmune destruction of insulin-producing pancreatic β cells, and the latter is caused by insulin resistance coupled with failure of β cells to compensate [1]. Type 2 DM (T2DM) is a public health problem that achieved pandemic extent because of its increased incidence rate. [88]. The reoccurrence of hyperglycemia may give rise to a variety of complications, such as neuropathy, retinopathy, and nephropathy, as well as increased risk of cardiovascular diseases [89,90]. Factors such as diet, inactive lifestyle, obesity, age, and genetics are reported to be responsible for the development of DM and its complications [91].
In the case of type 1 diabetes, insulin regulates the blood glucose level by signaling the cells to capture sugar from the bloodstream, but this uptake is interrupted by the destruction of β cells [92]. This is brought about by both the environmental and genetic factors under a stress environment [93]. Stress triggers the production of auto-antigens such as insulin, tyrosine phosphatase, glutamic acid decarboxylase, and islet cell antigen 69 from β cells [94,95]. These auto-antigens are recognized by the auto-antibodies and auto-reactive T cells that provide specificity to autoimmune destruction [96,97]. The infiltration of macrophages, dendritic cells, and CD4+ and CD8+ T cells, as well as the balance of CD4+ Th1/Th2, is responsible for the damage of β cells. Antigen-presenting cells, such as macrophages, present the antigen peptides to the circulating T cells through MHC molecules in the pancreatic lymph nodes, which activate the T cells. NF-κB is involved in the activation of the auto-reactive T cells, monocytes, and dendritic cells and is activated by pro-inflammatory cytokines [98]. The activated CD4+ Th1 T cells secret IL-2 and interferon-γ, which in turn activate the CD8+ T cells and other macrophages, which secrete various inflammatory cytokines, such as interleukin-1β (IL-1β), TNF-α, and ROS. These inflammatory cytokines play an important role in β-cell destruction [99,100]. Among them, IL-1β was reported to be the most destructive since it inhibits the mitochondrial function of the cell by generating NO [101]. In addition to these contributory factors, inflammation-driven DM development is gaining interest in the scientific community. It was found that inflammatory routes are involved in the progression of type 1 and type 2 DM; however, further studies suggest that different causal inflammatory pathways are associated with each type of DM.
The perception of association of inflammation with T2DM provides an exciting and new insight for a better understanding of the disease. Inflammation is involved in both the pathogenesis and the consequences of the disease. In diabetes, polyphenols were found to play an important role in suppressing blood glucose levels, enhancing the antioxidant status in pancreatic cells, and increasing the activation of PPARγ [102]. The probable mechanisms via which polyphenols effect this condition represent their hypocholesterolemic property, hypolipidemic activity, and antioxidant and free-radical scavenging properties. The association of inflammation in metabolic disease can be seen as altered upregulated expression of the pro-inflammatory cytokine TNF-α in adipocytes of obese animals. Binding of TNF-α with its soluble receptor results in a decrease in insulin resistance in obese animals. These findings link the bridge between elevated expression level and plasma concentration of pro-inflammatory cytokines in insulin resistance. Insulin resistance is an intricate metabolic condition, which causes insensitivity to insulin, as well as its downstream metabolic actions under normal serum glucose concentrations to the three metabolic tissues, i.e., liver, skeletal muscle, and white adipose tissue [103].
Insulin resistance plays a key role in metabolic syndromes and was also shown to be associated with nonalcoholic fatty liver disease [104]. Insulin shows its effect by binding to the surface of insulin-responsive cells and, as a result, phosphorylation occurs to the activated insulin receptor and other several substrates, including insulin receptor family member (IRS). This consequently initiates the downstream signaling cascades [105]. Inflammatory signaling targets this signaling pathway downstream of the insulin receptor, which leads to the insulin resistance. TNF-α exposure to cells or increased free fatty acid (FFA) stimulates the inhibitory phosphorylation of the serine residue of IRS-1. This inhibitory phosphorylation reduces the level of tyrosine phosphorylation of IRS-1 in response to insulin, as well as affects its binding ability to the insulin receptor, thus leading to the inhibition of downstream signaling and insulin effect.
Binding of TNF-α to its receptor activates several signaling pathways, which result in the activation of many transcription factors, such as NF-κB and JNK. As soon as these transcription factors are activated, they cause phosphorylation of IRS-1 at the 307th position (serine), and this results in insulin-receptor mediated tyrosine phosphorylation of IRS-1. A study on the human aortic endothelium showed that TNF-α regulated reduced tyrosine phosphorylation, as well as downregulated expression of the insulin receptor itself [106]. Recently, IL-6 was reported to inhibit insulin signal pathways in liver cells. This effect is related to suppressor of cytokine signaling-3 (SOCS-3), a protein which shows an association with the insulin receptor. IL-6 causes inhibition of SOCS-3 auto-phosphorylation, IRS-1 phosphorylation at its tyrosine residue, binding affinity between p85 subunit of phosphoinositide-3-kinase (PI3K) to IRS-1, and subsequently activation of Akt pathway. These effects of IL-6 are reported in both in vitro HepG-2 cells and in vivo mice [107].
IL-6, a pro-inflammatory cytokine, is released by a number of tissues, mainly adipose tissue, and it causes insulin resistance by downregulating the expression of glucose transporter-4 (GLUT-4) and IRS-1. The downregulated expression of GLUT-4 and IRS-1 is driven by IL-6-mediated activation of the JAK–STAT signaling pathway and increased level of SOCS-3 suppressor [108,109]. Thus, suppression of the level of serum IL-6 could ameliorate the insulin resistance complications [110]. Other mechanisms via which IL-6 causes insulin resistance are blockade of the PI3K pathway, impaired glycogen synthesis by decreasing the expression of miR-200s, and upregulation of friend of GATA-binding factor 2 [111,112,113]. Kim et al. [114] suggested that resistance of human skeletal muscle to insulin is IL-6-driven and leads to STAT-3-mediated TLR-4 gene expression. Insulin resistance causes a reduction in antilipolytic activity of insulin, which subsequently increases hepatic triglyceride synthesis [115]. Furthermore, the association of chronic inflammation to insulin resistance is also acknowledged. The responsible mechanisms via which chronic inflammation induces T2DM are not well understood and subject to further research. However, it was found that, in obese conditions, adipocytes synthesize and secrete pro-inflammatory cytokines (IL-1, IL-6, and TNF-α), and they are involved in several metabolic pathways related to insulin resistance, ROS production, lipoprotein lipase activity, and adipocyte function [116]. Therefore, both activated innate and acquired immunity play a vital role in the pathogenesis of diabetes, with convincing data that type 2 diabetes includes an inflammatory component [117].

2.3. Inflammation in Obesity

Obesity is at the center of the metabolic disorders, and it is associated with insulin resistance, CVD, atherosclerosis, type-2 diabetes, degenerative disorders, fatty liver disease, airway disease, and certain cancers [89]. Inflammation is commonly observed in obese and overweight patients. The inflammation induced by obesity is regarded as a low-grade chronic inflammation. [118,119]. As a result of inflammation, the adipose tissues release many inflammatory mediators. In many studies, the plasma concentrations of inflammatory biomarkers were found to be significantly higher than normal, non-obese subjects [120,121]. Macrophages and adipocytes secrete several proteins that modulate overall metabolic machinery, including fat storage [122,123]. The first molecular link between inflammation and obesity is TNF-α. It is still somewhat unclear what factors trigger the expression of TNF-α. However, previous documentations suggest that dietary fatty acids (FAs), such as long-chain, marine-derived n-3 FAs, elicit anti-atherogenic and ant-inflammatory effects [124]. It can be said that the level of TNF-α is nutritionally regulated and, more specifically, it can also be enhanced by hyperinsulinemia alone [125]. This inflammatory cytokine is predominantly secreted by monocytes and macrophages. The activation of the TNF receptor results in stimulation of NF-κB signaling via inhibitor of κB kinase β (Ikkβ) [126]. The NF-κB/Rel family includes NF-κB1 (p50/p105), NF-κB2 (p52/p100), p65 (RelA), RelB, and c-Rel [127]. These molecules represent a family of transcription factors that are normally found in the cytoplasm in an inactive state. They are associated with a regulatory protein from the inhibitors of κB (IkB) protein family, which includes IκBγ, IκBβ, IκBα, IκBε, and Bcl-3 in higher vertebrates [128]. In response to the multiple stimuli of inflammatory cytokines, the IκBα, which is bound to the p50–p56 heterodimer and the p50 homodimer, is phosphorylated by IKK. The IKK complex consists of at least three subunits, namely, kinases IKKα, IKKβ, and IKKγ [127,129]. IKK phosphorylates IκBα specifically at the NH-terminal serine residue, which is then consecutively ubiquitinated and selectively degraded by the 26S proteasome, thereby releasing NF-κB [130]. The free NF-κB then binds to κB enhancer elements of target genes and induces transcription of pro-inflammatory genes [127,131].
In addition, the contribution of a number of mediators was suggested. The adipokines such as leptin, adiponectin, IL-6, and TNF-α show prominent effects on adipocyte metabolism and utilization of insulin and, therefore, exhibit a strong association with obesity and related metabolic disease [132]. However, the inflammation induced by obesity is low-grade and primarily supported by low concentrations of inflammatory cytokines. It was observed that obesity-induced inflammation exhibits similar characteristics in molecular aspects to atherosclerosis, which is counted as one of the major complications of metabolic syndrome in line with lipid metabolism disorder and insulin resistance [133,134,135]. It is worth mentioning that the reported findings clearly demonstrate the involvement and significance of immune cells in obesity-induced inflammation.

2.4. Inflammation in Fatty Liver Disease

Fatty liver disease, also identified as hepatic steatosis, is caused by the accumulation of various fatty acids, mainly triglycerides, in the liver [136]. FFAs are the predominant type of lipids in the liver (nearly two-thirds of lipids). The accumulation of lipids in the liver is known to result from elevated de novo hepatic lipogenesis, lipolysis from visceral adipose tissue, decreased secretion of lipoprotein triglycerides, and free fatty oxidation [137].
Lipid aggregation and a stressful environment in the mitochondria of liver cells lead to the production of TNF-α and ROS, which further contribute to inflammation [138]. Inflammation is an integral part of nearly all acute and chronic liver disorders including fatty liver disorders, such as alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD). Pro-inflammatory cytokines regulate important features of liver disorders, including acute phase response, acute liver failure, cholestasis, hypergammaglobulinemia, steatosis, and fibrosis development [139,140]. The acute phase response is critically regulated by inflammatory cytokines. One of the special characteristics of many chronic liver disorders is sterilized inflammation, especially ALD and NAFLD [141]. Inflammatory cytokines, e.g., IL-1family members (IL-1α, IL-1β, IL-1Rα, IL-18, IL-33, IL-36, IL-37, and IL-38), are associated with the regulation of atherosclerosis, insulin resistance, and adipose tissue inflammation, all of which are common features of NAFLD [142,143].
There are several adipose tissue-derived signals that are necessary for the prevention of inflammation and for proper functioning of the liver. Obesity and insulin resistance are both responsible for lipid accumulation and liver inflammation [144]. Expression of the two most important pro-inflammatory cytokines, TNF-α and IL-6, is enhanced by the elevation of fat and insulin resistance in the body [145]. The most affected cellular organelle in this hepatic steatosis disease is the endoplasmic reticulum, whose activation is directly linked to an insulin-resistant state [146]. Many factors, such as the abnormal concentration of ROS, imbalance of Ca2+, and accumulation of unfolded proteins, lead to the activation of endonuclease inositol-requiring protein 1 [147]. The stimulation of the protein kinase and the secretion of FFAs from adipose tissue induce JNKs or stress-activated protein kinases (SAPKs). JNK stimulation is mediated by diverse factors, including TNF-α, IL-1, epidermal growth factor (EGF), tumor growth factor-β (TGF-β), and ROS [148]. The activated JNK signals macrophage accumulation, normal T-cell expression, and secretion of AP-1 and IL-8, which further contribute to the inflammatory conditions [149]. The endoplasmic reticulum (ER) oxidative stress directly induces the activation of IκB kinase, while MAP kinase, like NF-κB-inducing kinase, plays an indirect role. The phosphorylation of IKKα and IKKβ stimulates the expression of NF-κB and furthers the NF-κB p65–p50 heterodimeric complex. The complex interacts with the inflammatory cascades via the transcription of pro-inflammatory molecules, such as NO, adhesion molecules, cyclooxygenases (COXs), cytokines (IL-6, IL-1β, TGF-β, and TNF-α), and chemokines [150].

2.4.1. Acute Alcoholic Hepatitis (AAH)

Alcoholic liver disease (ALD) is characterized by diverse metabolic features, including neutrophilia, anorexia, fever, muscle catabolism, activation of monocytes and macrophages, and altered mineral metabolism. IL-1 and TNF-α were shown to be involved in most of these processes [142]. Acute alcoholic hepatitis (AAH) was the first disease characterized by an increase in serum IL-1 activity. Fever and neutrophilia are AAH’s distinctive clinical features [151]. Patients with severe AAH are reported to have very high (almost 10-fold) serum IL-1 activity. AAH is among the first few diseases in which elevated levels of TNF-α were detected [152]. Blood levels of TNF-α and soluble tumor necrosis factor receptors are directly correlated with endotoxemia, impaired intestinal permeability, and mortality. IL-8 expression in serum is under the direct control of IL-1 and TNF-α. Hence, serum IL-8 levels are correlated with the diagnosis of AAH patients. Since IL-8 is responsible for neutrophil recruitment into the liver, its hepatic expression correlates with the progression of patients with alcoholic hepatitis (AH) [153]. ALD patients show increased levels of pro-inflammatory cytokine IL-18 in the serum, as well as in peripheral blood mononuclear cells. Higher levels of serum IL-18 and its natural antagonist, IL-18 binding protein (IL-18BP), are related to mortality in AAH [154]. An elevated level of IL-17, another inflammatory cytokine that affects neutrophil recruitment, is also observed in human ALD. Its level is directly correlated with liver inflammation [155]. Research on ALD revealed that AAH is a cytokine-driven disorder, characterized by the detectable quantity of many pro-inflammatory mediators in the systemic circulation Therefore, targeting the cytokine pathway could be a better strategy for therapeutic intervention [139].

2.4.2. NAFLD

NAFLD can be defined as a collection of fat (>5%) in hepatocytes without the intake of excessive alcohol [156,157]. More than 30% of the population of the Western world is affected by this disease, particularly obese (76%) and T2DM (50%) patients. Pathogenesis of NAFLD is complex and exerts its harmful effect on liver cells via two mechanisms, i.e., accumulation of fatty acid in liver cells and oxidative stress-mediated damage to hepatocytes [158].
Accumulation of triglycerides in liver cells occurs either due to excessive intake of saturated fatty acids and obesity or because of hyperglycemia and hyperinsulinemia as in the case of insulin resistance [159]. Hyperglycemia and hyperinsulinemia upregulate the expression of carbohydrate response element-binding protein (ChREBP) and sterol regulatory element-binding protein (SREBP-1c), respectively. This in turn activates the expression of genes associated with the FFA synthesis and also decreases β-oxidation of fatty acid [160]. Inflammatory damage to liver cells results in the increased expression of PPAR-γ, whose expression leads to the accumulation of FFA in the liver. Liver X receptor (LXR) upregulates the expression of SREBP-1c and ChREBP, which are genes involved in the FFA synthesis that leads to steatosis [161].
Oxidative stress is the second mechanism via which NAFLD harms the liver. During liver inflammation, hepatocytes and inflammatory cells secrete cytokines, such as IL-6, TNF-α, and ROS. [158]. All of these activities activate hepatic endothelial cells, which increase cytokine expression and finally activate hepatic stellate cells, which cause phenotypic changes related to pro-fibrinogenic and pro-inflammatory functions [162].
Excessive accumulation of lipids leads to hepatocyte injury and activates an inflammatory response, increasing the risk of liver disease [163]. The immune response of the liver is formed by immune cells such as dendritic cells (DCs), natural killer cells (NK), Kupffer cells, neutrophils, monocytes, and NK T cells (NKT). These immune cells commence and sustain hepatic inflammation via an assembly of chemokines and cytokines, especially IL -1β and TNF, as well as ROS [164].
The activation of hepatic inflammation, as mentioned above, is caused by the build-up of infectious and non-infectious material, which is produced during cell injury and is predicted by pattern recognition receptors (PRRs). These PRRs include NLRs, C-type lectin receptors (CLRs), TLRs, and several other receptors [165]. In the pathological condition of NAFLD, the accumulation of fatty acids causes inflammatory cascades in the hepatocytes, which results in caspase-1 activation, as well as the production of TNF-α and IL-1β. Among the different NLRs, the NLRP3 inflammasome is known as a key contributor to the pathological development of inflammatory-associated diseases. The NLRP3 inflammasome is involved in the activation of apoptosis-associated speck like protein to CARD (ASC), which can result in the release of proinflammatory cytokines, such as IL-1β and IL-18. This leads to the development of NAFLD [158,166].

3. Flavonoids as Anti-Inflammatory Agents in Treating Metabolic Disorders

The global flavonoids market demands were estimated to be $840.2 million in 2015 and predicted to reach above $1.06 billion in 2025 [167]. Owing to flavonoids’ availability, safety, and low cost, as well as their considerable antioxidant and anti-inflammatory activities, in addition to their wide usage in functional foods, beverages, and dietary supplements, this market is expected to grow steadily. Flavonoids represent the largest class of polyphenols, which are the most abundant plant-derived bioactive compounds. The flavonoid chemical structure presents a peculiar C6–C3–C6 backbone structure [168]. In particular, it consists of two aromatic rings (also called A and B rings) that are linked by a three-carbon-chain, generating an oxygenated heterocycle (C ring) [51]. On the basis of their heterocycle structure, flavonoids are divided into several classes, including flavanols ((−)-cathecin, (+)-gallocatechin, (−)-epicatechin, (−)-epigallocatechin, (−)-epigallocatechingallate, epigallocatechin gallate, theaflavine, and theaflavinegallate), flavones (apigenin and luteonin), flavonols (kaempherol, myricetin, quercetin, isorhemetin, and rutin), flavanones (eriodictyol, hesperidin, and naringenin), anthocyanidins (cyanidin, delphinidin, malvidin, pelargonidin, peonidin, and petunidin), and isoflavones (genistein) [51,169,170,171]. Figure 2 presents chemical structures of some of the most relevant flavonoids belonging to the aforementioned classes.
Flavonoids are ubiquitously present in plants [51,172], including several plant-based foods [51,168,170], where they mainly exist in a glucoside form [172,173] and, for this reason, they are poorly absorbed [172]. Flavonoids are widely present in plant-based diets, such as the Mediterranean diet, which is characterized by a large and daily consumption of fruits, vegetables, whole grains, extra-virgin olive oil, herbs and spices, and red wine [174]. Among plant-based foods, onion, leeks, curly kale, broccoli, blueberries, and red wine are the richest sources of flavonoids [168]. In addition, apples, grapes, citrus fruits, tea, red pepper, and cocoa are other flavonoid-rich substances [170].
The most important action of these compounds is their antioxidant activity, mainly exerted by chelating metal ions or by free-radical scavenging due to the flavonoid hydroxyl group [172], thus suggesting their pivotal role in prevention and management of several pathologies including CVD, cancer, diabetes, and chronic-degenerative diseases [51], which are induced by persistent oxidative stress [175]. Interestingly, oxidative stress, in particular the endogenous production of oxidative compounds such as ROS and RNS, is strongly related to an inflammatory status [51,175,176]. In addition, a chronic inflammatory status was recognized as a major risk factor for the pathogenesis of several metabolic disorders, including CVD and diabetes [13]. Taking into account their activity against oxidative stress, dietary antioxidants might also be considered good candidates for the management of inflammation.
The anti-inflammatory activity of polyphenols, specifically flavonoids, was evaluated by both in vitro and in vivo analysis. Although there are a number of anti-inflammatory targets and mechanisms of flavonoids, the most important is the inhibition of eicosanoid-generating enzymes, including COXs, phospholipase A2, and lipoxygenases, which decrease the concentration of prostanoids and leukotrienes [177]. Other mechanistic targets include histamine release inhibition, phosphodiesterase, protein kinases, and activation of transcriptase. Inhibition of the COX cascade is observed with the in vitro treatment of flavonoids, such as quercetin. Quercetin is a strong inhibitor of COX-2 and 5-lipoxygenase (5-LOX), which are both involved in the production of eicosanoids from arachidonic acid [178]. It was also found that citrus polymethoxy flavones inhibited the production of cytokines, TNF-α, macrophage inflammatory protein-1, and IL-10 via the activated monocytes [179]. These flavones also furthered the activation of phase II antioxidant enzyme MAP kinase, protein kinase C (PKC), and Nrf2 activity [170].
The inhibition pathway of NF-κB was widely studied using a number of flavonoids, such as fisetin, silymarin, quercetin, kaempferol, rutin, luteolin, apigenin, isoliquiritigenin, xanthohumol, and chrysin. It was found that these flavonoids inhibited NF-κB activation by inhibiting a particular step in the NF-κB activation pathway. It is known that NF-κB is found in the cytoplasm associated with the IκB protein. Upon receiving signals, certain genes such as NF-κB-inducing kinase (NIK), mitogen activated protein kinase kinase (MEKK), interleukin-1 receptor-associated kinase (IRAK), TNF receptor-associated factor (TRAF), PKC, and VCAM lead to the activation of IKK. In the presence of various flavonoids, the activation of the aforementioned genes can be inhibited or mediated to inhibit the activation of IKK. Some flavonoid, such as fisetin and apigenin, can inhibit the IKK complex formation. Silymarin, quercetin, and isoliquiritigenin can inhibit activation of NF-κB transcription. Isoliquiritigenin can also inhibit NF-κB–IκB complex formation. Morin and rutin inhibit IκB, while apigenin, silymarin, kaempferol, and isoliquiritigenin inhibit phosphorylation of IκBα. Ubiquitination of IκB via the ubiquitine ligase system is inhibited by apigenin. The degradation of IκB by 26S proteasome (26S) is inhibited by quercetin and isoliquiritigenin. Translocation of activated NF-κB into the nucleus can be inhibited by isoliquiritigenin. Finally, the interaction of NF-κB with the κB binding sequence to enhance NF-κB-regulated genes may be inhibited by apigenin [180]. Similarly, various MAP kinase or JNK pathways are regulated by flavonoids which control inflammation by inhibiting Jun/ AP-1 or activation of Nrf2 and Kruppel-like factor 2 [181,182].
In some aspects, flavonoids seem to act through mechanisms similar to those of some anti-inflammatory drugs. In addition, flavonoids are able to inhibit aggregation and adhesion of platelets [183]. Interestingly, flavonoids were also demonstrated to be effective in inhibiting poly(ADP-ribose) polymerase 1 (PARP-1) [184,185], which is strongly involved in acute and chronic inflammation by acting as an upregulator in several pro-inflammatory pathways [186]. Further in vitro evidence showed a marked effect of flavonoids in suppressing several inflammatory biomarkers’ levels [187,188]. In this context, various flavonoids are reported to exert anti-inflammatory activity, including quercetin, kaempferol, catechins, morin, myricetin, apigenin, luteolin, genistein, silybin, and hesperidin [51,170].

3.1. In Vitro Studies on Flavonoids as Anti-Inflammatory Agents in Treating Metabolic Disorders

Although in vitro evidence showed the anti-inflammatory potential of flavonoids [189], human clinical trials are scarce and provide contrasting evidence [170]. This is probably due to the study design used or the outcomes evaluated [173,176]. Interestingly, in 2011, a cross-sectional study was conducted on a large number of healthy women from the Nurses’ Health Study cohort in order to investigate the effects of flavonoids on inflammation and endothelial function markers [190]. In particular, flavonoid intake was assessed by using a food frequency questionnaire and divided into six subclasses (flavones, flavan-3-ols, flavonols, anthocyanidins, polymeric flavonoids, and flavanones). The first significant result obtained was an inverse correlation between several classes of flavonoids and inflammatory markers. Particularly, after multivariable adjustment, a statically significant association was found between IL-18 and flavones, anthocyanidins, and total flavonoids (−0.069, p = 0.033; −0.079, p = 0.014; and −0.075, p = 0.020; respectively), VCAM-1 and flavonols (−0.078, p = 0.012), and tumor necrosis factor receptor (TNF-R)2 and flavanones (−0.061, p = 0.05) [190]. Similarly, Steptoe demonstrated that chronic consumption of tea significantly decreased CRP when compared to placebo (p = 0.05) [191].
It is well known that the anti-inflammatory activities of natural bioactive compounds, including flavonoids, play a pivotal role in the management of CVD [192]. A recent narrative review summarized the available literature about the effects of flavonoids in CVD and highlighted their action on several molecular targets involved in CVD, including reduction in the expression of cytokines, adhesion molecules, and other inflammation markers, such as CRP [193].
Many studies investigated the flavonoids anti-inflammatory activity. In an in vitro study carried out on human aortic endothelial cells, it was shown that flavonoids, in particular flavanols (including kaempferol, quercetin, and galangin) and hydroxyl flavones (including apigenin and chrysin), were effective in inhibiting the expression of endothelial adhesion molecules induced by TNF-α, such as intercellular adhesion molecule 1 (ICAM-1) and E-selectin [194]. Additionally, in vitro evidence also reported that quercetin was effective in reducing the transcription factor activator protein (AP)-1, resulting in decreased expression of ICAM-1 [195]. Further evidence showed that proanthocyanidins, from grape seed, acted by downregulating TNF-α-induced expression of VCAM-1 [196]. Moreover, genistein, one of the most abundant isoflavones, was reported to be able to inhibit cytokine-induced adhesion of monocytes in human endothelial cells at physiological concentrations [197,198]. This suggests a marked anti-inflammatory mechanism of isoflavones in protection against vascular disease. Similar evidence was found during a study on animal models, in which administration of soy isoflavones was reported to be effective in reducing the risk of inflammation-related CVD by acting through TNF-α downregulation at the endothelial level [199].
The effects of flavonoids from cocoa on inflammatory markers were investigated in an in vitro study. In particular, the cellular effects of cocoa extract (CE), epicatechin (EC), and isoquercitrin (IQ) in equivalent concentrations were compared. CE, EC, and IQ reduced TNF-α levels in a dose-dependent manner, but the CE effect was higher than that produced by EC and lower than that produced by IQ. In addition, CE and EC were found to be effective in reducing the mRNA expression levels of TNF-α, IL-1α, and IL-6 [200] (Table 1).

3.2. In Vivo Studies on Flavonoids as Anti-Inflammatory Agents in Treating Metabolic Disorders

Zhu et al. [228] showed the anti-inflammatory effect of anthocyanins based on in vitro and in vivo studies. In particular, the treatment with an anthocyanin mixture (50 mg per mL) significantly decreased the production of CRP induced by IL-6 and IL-1β in HepG2 cells (p < 0.05) and the lipopolysaccharide (LPS)-induced expression levels of VCAM-1 in porcine iliac artery endothelial cells (PIECs) (p < 0.05). In addition, a 24-week administration of a purified anthocyanin mixture (320 mg per day) in moderate hypercholesterolemic subjects significantly reduced CRP (p < 0.001), VCAM-1 (p = 0.005), and IL-1β (p = 0.019) serum levels when compared to placebo [203].
In order to investigate the effects of flavonoids on human health, nutritional intervention-based clinical trials were conducted using specific foods naturally rich in flavonoids or enriched with these compounds. Among these, cocoa or chocolate are good candidates due to their high flavonoid content [229,230] and because their intake can be considered as an easy intervention in clinical trials. In a randomized, cross-over, placebo-controlled trial, it was observed that four-week consumption of 15 g of cocoa product significantly reduced the levels of IL-10 and IL-1β (p = 0.001) in moderate hypercholesterolemic subjects [231]. According to the authors, this anti-inflammatory effect was attributed to flavonoids contained in cocoa [232]. Similarly, flavonoids from cocoa powder (40 g per day for four weeks) significantly reduced p-selectin and ICAM-1 (both p = 0.007) in high-CVD-risk subjects [233]. Similar results were also found by Wang-Polagruto and colleagues (2006), which showed that a six-week administration of 446 mg of total flavanols from cocoa significantly reduced VCAM-1 levels in hypercholesterolemic postmenopausal women [234].
An interesting pilot randomized controlled study was conducted on obese and diabetic subjects in order to investigate the effects of chocolate polyphenols on endothelial function after induced hyperglycemia [235]. In particular, 13.5 g of high-polyphenol (HP) or low-polyphenol (LP) (control) chocolate was administrated after 12 h of fasting. One hour after chocolate intake, a glucose load (75 g) was given to induce hyperglycemia, and then inflammatory markers were assessed 120 min after. Authors found that HP, when compared to control, showed reduced ICAM-1 (ng/mL, HP vs. LP; fasting: 325.6 ± 9.0, after glucose load: 310.0 ± 8.4 (p = 0.20) vs. fasting: 321.1 ± 7.6, after glucose load: 373.6 ± 10.5 (p = 0.04)), p-selectin (ng/mL, HP vs. LP; fasting: 253.0 ± 14.8, after glucose load: 235.0 ± 7.7 (p = 0.62) vs. fasting: 265.0 ± 15.2, after glucose load: 268.5 ± 12.4 (p = 0.92)), E-selectin (ng/mL, HP vs. LP; fasting: 111.3 ± 5.8, after glucose load: 96.6 ± 5.6 (p = 0.09) vs. fasting: 94.4 ± 4.0, after glucose load: 105.8 ± 3.5 (p = 0.28)), and p-selectin glycoprotein ligand 1 (U/mL, HP vs. LP; fasting: 281.9 ± 12.2, after glucose load: 212.6 ± 8.7 (p = 0.13) vs. fasting: 262.9 ± 5.5, after glucose load: 327.5 ± 7.3 (p = 0.09)) [235]. Although this pilot study was conducted on a small number of subjects, it provides evidence about the acute beneficial effects of flavonoid intake, from a dietary source, on inflammation.
In a randomized, cross-over, double-blind, placebo-controlled clinical trial, Dower and colleagues [236] investigated the effects of flavonoids on inflammation and endothelial dysfunction markers on pre-hypertensive subjects. Equimolecular amounts (345 µmol) of quercetin and epicatechin were administrated for four weeks. The serum levels of VCAM-1, ICAM-1, E-selectin, von Willebrand factor (vWf), monocyte chemotactic protein (MCP)-1 (endothelial dysfunction biomarkers), IL-1β, IL-6, IL-8, TNF-α, CRP, and serum amyloid A (SAA) (inflammation biomarkers) were evaluated. All biomarkers’ levels decreased after epicatechin and quercetin treatment, and statistically significant decreases were found in E-selectin levels by epicatechin (−7.7 ng/mL; 95% confidence interval (CI): −14.5, −0.83; p = 0.03) and in E-selectin and IL-1β by quercetin (−7.4 ng/mL; 95% CI: −14.3, −0.56; p = 0.03 and −0.23 pg/mL; 95% CI: −0.4, −0.06; p = 0.009, respectively) [237].
The anti-inflammatory effects of the flavonoids from red wine were highlighted in a randomized, cross-over study in which 100 mL of red or white wine was administered twice daily (20 g ethanol). After four weeks of intervention, the authors found that, in both the red and white wine groups CRP, ICAM-1, and IL-6 levels were significantly reduced (p < 0.01); however, VCAM-1 and E-selectin levels were significantly decreased only in the red wine group (p < 0.01), suggesting a greater anti-inflammatory effect of red wine due to its high polyphenol content [238]. Interestingly, a one-month administration of 100 g per day of grape seed-derived proanthocyanidins was reported to be able to reduce the levels of VCAM-1, ICAM-1, and selectin in patients with systemic sclerosis [239]. This shows the anti-inflammatory activity of this class of flavonoids.
Karlsen and colleagues [240] conducted a randomized, placebo-controlled trial to investigate the effect of bilberry juice on inflammatory biomarkers in subjects with elevated risk of CVD. They found statistically significant reduced levels of CRP, IL-6, IL-15, and monokine induced by interferon (INF)-γ in the treatment group compared to placebo (p = 0.027, p = 0.037, p = 0.008, and p = 0.047, respectively) [240]. These anti-inflammatory effects were attributed to flavonoids contained in the bilberry juice, which is particularly rich in anthocyanins.
Strawberries are another flavonoids-rich dietary source. In particular, flavonols, flavanols, anthocyanins, tannins, and hydroxycinnamic acid derivatives are contained within strawberries [241]. The effect of a strawberry beverage on post-prandial inflammatory markers was investigated in overweight and obese (body mass index (BMI) ranged from 25 to 33.5 kg/m2) subjects [242]. After a seven-day run-in period, subjects consumed high-energy, high-carbohydrate, and moderate-fat test meals, accompanied with strawberry or strawberry-flavored (placebo) beverages, in random order. The blood levels of glucose, insulin, CRP, IL-6, IL-1β, plasminogen activator inhibitor (PAI)1, and TNF-α were monitored. Blood levels of anthocyanins were used as an intake marker. The authors found that, six hours after the test meal, strawberry significantly decreased the levels of CRP (3.1 ± 0.1 and 2.7 ± 0.1 mg/L, in treatment and placebo group, respectively; p = 0.02), IL-6 (3.1 ± 0.2 and 2.6 ± 0.2 ng/L, in treatment and placebo group, respectively; p = 0.05), and insulin (458.4 ± 13.9 and 402.8 ± 13.9 pmol/L, in treatment and placebo group, respectively; p = 0.01) [242].
It is well known that high blood glucose levels are one of the most important cardiovascular risk factors. In particular, evidence showed that periodic hyperglycemia strongly impairs the endothelial function through increasing oxidative stress [243]. Persistent oxidative stress, in turn, seems to be responsible for the impairment of β-cell function, suggesting its pivotal role in the pathogenesis of T2DM [228,244,245].
Hyperglycemia-induced oxidative stress also acutely increases pro-inflammatory cytokine levels [246]. In addition, insulin-resistance (IR) was reported to be strongly related to inflammation [247,248], particularly in obesity and T2DM [247]. Moreover, obesity is characterized by a chronic low-grade inflammatory status, which is recognized as one of the main causes of IR [249]. Furthermore, IR and inflammation are also involved in NAFLD, forming a vicious circle with conditions promoting this disease process [250]. In this context, the effects of flavonoids in reducing oxidative stress and/or improving IR might be considered as an interesting strategy in ameliorating inflammatory status in metabolic disorders.
Recent systematic reviews and meta-analyses focused on the effects of flavonoids on various metabolic risk factors, including IR [236,251,252]. In particular, evidence from randomized clinical trials (RCTs) showed that isoflavones from soy are able to reduce insulin (−1.37; 95% CI, −1.92, −0.81) and IR when evaluated with the Homeostatic Model Assessment (HOMA) (−0.39; 95% CI, −0.62, −0.14) [251]. Shrime et al. (2011) showed that, in short-term studies, flavonoid-rich cocoa is effective in reducing HOMA (−0.94 points; p < 0.001) [252]. Similarly, Hooper et al. (2012) provided evidence that flavan-3-ols are able to reduce homeostasis model assessment of insulin resistance (HOMA-IR) (−0.67; 95% CI, −0.98, −0.36), in RCTs [236].
A double-blind, placebo-controlled RCT was carried out on postmenopausal diabetic women in order to examine the effects of flavonoids on cardiovascular risk factors, including IR. The nutritional intervention consisted of an intake of 13.5 g of flavonoid-enriched chocolate twice daily, providing 100 mg of isoflavones and 850 mg of total flavan-3-ols. Adherence was monitored by the assessment of urinary levels of flavan-3-ols and isoflavones. After 12 months, a significant reduction in IR (HOMA-IR: −0.3 ± 0.2; p = 0.004) was found in the intervention group when compared to placebo [253]. Further in vivo studies were conducted in order to investigate the effects of flavonoids from cocoa on IR. In particular, the authors showed that daily consumption of 100 g of dark chocolate (containing about 500 mg of polyphenols and, more specifically, 65.97 mg of epicatechin, 21.9 mg of catechin, 0.59 mg of quercetin, 0.31 mg of isorhamnetin, and 0.03 mg of kaempferol) is able to significantly reduce HOMA-IR (p < 0.001) and increase insulin sensitivity (p < 0.001) when assessed by the Quantitative Insulin Sensitivity Check Index (QUICKI) (both calculated by oral glucose tolerance test (OGTT)) and compared to 90 g of white polyphenol-free chocolate [254,255]. Desideri et al. (2012) conducted a double-blind, parallel arm study in 90 elderly subjects, evaluating the effects of flavanols on various biological markers, including HOMA-IR. Subjects were randomized in three groups: high (HF), intermediate (IF), and low flavanol consumption (LF), consisting of ~990 mg, ~520 mg, and ~45 mg per day, respectively. After an eight-week treatment, HOMA-IR was significantly reduced in a dose-dependent manner in the HF and IF groups (−1.6 ± 1.0; p < 0.0001 and −0.9 ± 0.2; p < 0.0001, respectively) but not in the LF group (−0.1 ± 0.5; p = 0.29), suggesting that high consumption of dietary flavonoids is effective in improving insulin sensitivity [256].
Weseler and colleagues [257] conducted an ex vivo study investigating the effects of flavonoids on inflammatory markers. In particular, blood from T2DM subjects was incubated with 10 µmol/L flavonoids (flavone, fisetin, tricetin and morin) and 30 min after cytokine release was stimulated by treatment with LPS. After 16 h of incubation, tricetin and fisetin significantly reduced IL-6 levels (− 29 ± 6% and −31 ± 5%, respectively; p ≤ 0.001 [257].

4. Clinical Trials of Flavonoids in Metabolic Disorders

Flavonoids are a widespread group possessing the phenylchromane moiety. Depending on the substitution pattern, they can be subdivided into flavonols, flavanols, flavones, flavanones, isoflavones, and anthocyanins [258]. Despite flavonoids being renowned for their antioxidant capacity/activity, they have many other capabilities. Because of this fact, flavonoids can offer health benefits possibly derived from their antioxidant properties [259]; however, several studies indicated that their nutritional importance can possibly be linked to their anti-inflammatory action. Such benefits and properties can be associated with their roles in metabolic disorders and their risk factors. However, their impact requires the examination of a multifaceted scenario; thus, basic research and clinical trials need to be conducted to assess their role in the prevention/amelioration of such diseases’ end-points [258,260,261]. Although the evidence of the therapeutic uses and/or clinical efficacy of flavonoids in metabolic disorder-associated diseases is suggestive, the lack of satisfactory clinical verification makes those results significantly limited.
However, the current literature has some important examples of clinical trials on metabolic disorder-related diseases, such as insulin resistance, dyslipidemia, hypertension, and obesity [34,260]. Most of these studies are related to flavonoid-rich materials and supplements, which indicated good results in such disorders at different levels [176]. These products are regularly abundant in the diet and seem to be non-harmful in low to moderate doses. For instance, well-known flavonoid-rich supplements and foods were demonstrated to exhibit health benefits for cardiovascular disease risks, diabetes, and obesity in randomized controlled trials using green tea [31], black tea [262], cocoa [263,264], soy [265], cranberries [266], grape [267], olive [268], and silymarin [269], among others. Those studies evidenced that long- or short-term regular ingestion of the abovementioned supplements can improve/ameliorate/influence disease conditions when compared to controls. However, no specific flavonoid source can be considered superior to another, nor can the suitability of the use of a particular flavonoid source be considered more successful or reliable when it comes to health benefits. Clinical evidence for the effect of the isolated/purified flavonoid-like active principles is still lacking. Thus, there is a clear limitation to the advance of understanding with regard to the consistent effects, efficiency, and safety of these bioactive compounds on human metabolic disorders after consumption.
In this context, the effect of some isolated flavonoids was also evaluated in various clinical trials. In some cases, their efficacy in such diseases was confirmed, with evidence about the important impacts on metabolic disorder-related abnormalities. However, the information/results/conclusions are conflicting even in their design, temporality, doses, and parameters assessed. There are many different types of flavonoids found in functional foods, and several clinical trial results, using different flavonoids, are presented in Table 2. In this regard, quercetin, a naturally occurring flavonol, which is found in many food products (e.g., vegetables, leaves, fruits, and grains) as the major dietary flavonoid, was evaluated in various clinical trials. Despite the promising experimental findings at in vitro and preclinical levels, randomized controlled trials resulted in mixed outcomes regarding the impact of quercetin in metabolic disorders. In fact, some trials did not suggest any clinically relevant effects of quercetin supplementation. For instance, one study showed a reduction of plasma lipid levels [270], whereas other studies indicated reasonable clinical evidence for cardioprotection by anti-inflammatory means [271]. Those studies were designed as placebo-controlled, double-blind, randomized, cross-over trials in healthy or condition-having (e.g., prehypertensive, hypertensive, obese/overweight, dyslipidemic, postmenopausal, with rheumatoid arthritis, with sarcoidosis, with metabolic syndrome light-chain (AL) amyloidosis) male/female subjects (18 to 200). The evaluating doses of quercetin ranged from 150–2000 mg/day for one day to eight weeks. Results from these trials indicated some improvement in risk factors of CVD, even as cardioprotective in CVD episodes, through recovery of endothelial function and inflammation reduction [237,271,272,273]. However, in spite of the supra-nutritional doses employed, quercetin seems to have no effects on systemic and adipose tissue inflammation, as well as insulin and glucose levels; however, it reduces elevated plasma uric acid concentrations [274,275]. Thus, no influence on innate immune function or inflammation was perceived [276]. On the other hand, it can exert slightly pro-inflammatory effects [272]. Additionally, quercetin exhibited blood pressure-lowering effects in overweight–obese carriers of the apoE3 genotype and reduced the HDL cholesterol and apoA1 levels in apoE4 subjects [277], as well as improved the symptoms and disease activity in women with rheumatoid arthritis [278]. In the case of sarcoidosis and dyslipidemia, quercetin intake was related to a decrease in oxidative stress and inflammation in sarcoidosis, as well as positive effects on blood lipids [273,279].
Similar trends were documented for other type of flavonoids, such as the tea flavan-3-ols epicatechin and epigallocatechin-3-gallate (EGCG). The clinical evidence (Table 2) suggests a cardioprotective effect by increasing endothelial function [237], involving specific activation of associated downstream signaling pathways and antioxidant potential as the main bioactive action [280]. Among those cross-over, double-blind, placebo-controlled, randomized studies, doses of these flavanols were found to be in the 100–1890 mg/day range, between four and 24 weeks, and in conditioned male/female patients (Table 2). Analogously, citrus glycosylated flavanones (such as hesperidin and naringin) were clinically demonstrated to diminish the plasma cholesterol in hypercholesterolemic patients, as well as improve levels of different factors in type 2 diabetes, but no effect was evidenced on blood total and LDL cholesterol (LDL-C) concentrations [281,282]. These cross-over, double-blind, placebo-controlled, randomized trials were performed using doses ranging from 400–800 mg/day in conditioned subjects.
Placebo-controlled, double-blind, randomized, cross-over studies in conditioned patients (19–138) that took place during two weeks to three years of supplementation showed that the isoflavones genistein (54 mg/day) and puerarin (500 mg/day) can improve surrogate end-points associated with risk for diabetes and cardiovascular problems. This was found even in conditions such as postmenopausal women with metabolic syndrome [283] and acute myocardial infarction or coronary heart disease [284,285]. Furthermore, genistein also enhanced insulin sensitivity indexes in patients with hyperinsulinemia [286] and showed positive effects on some cardiovascular risk factors in postmenopausal women [287]. On the other hand, clinical efficacy was not detected during intake of genistein in patients with Sanfilippo disease [288].

5. Concluding Remarks and Future Perspectives

The inflammatory response is initiated in response to injury and infection. According to our limited understanding on the biological intricacy of chronic inflammatory disorders, the development of anti-inflammatory therapy remains a critical challenge. It is well known that oxidative stress and inflammation elicit generation of ROS and RNS, which trigger redox-sensitive kinases, such as ASK1, which in turn activate downstream MAPKs, NF-κB, and AP-1. This results in the induction of inflammatory gene expression. Polyphenolic compounds increase the level of anti-inflammatory genes, such as SOD, GPx, and HO-1 via activation of Nrf2 (Figure 3). Involvement of NF-κB, a well-known inflammatory and immunological mediator, was revealed in three common metabolic disorders, i.e., atherosclerosis, insulin resistance, and obesity. Flavonoids suppress inflammatory cytokines, modulate transcription factors and inflammation-related pathways, reduce the level of ROS by metal ion chelating or by radical scavenging, and inhibit eicosanoid-generating enzymes, including COXs, lipoxygenases A2, and phospholipase, thereby diminishing the concentration of prostanoids and leukotrienes. In conclusion, the current clinical synthetic drugs are not producing satisfactory results in the management of metabolic disorders induced by inflammation and are only effective in treating the disease-associated symptoms. Thus, new effective therapeutic agents that have the ability to encounter the pathology and restore the normal physiology are needed. Furthermore, experimental outcomes for several metabolic disorders also appear to vary in results and conclusions, limiting the applicability of those results to practice. However, some clinical findings indicated that quercetin and genistein seem to have cardioprotective effects and that flavanols have influences on inflammation; however, outcomes are too preliminary to be included within clinical practice. Due to this fact, further research that includes more rigorous, randomized, cross-over, placebo-controlled trials, based on a large number of patients/samples/data, and comprising both men and women, are required in order to provide clarification on affirmations regarding the healthy impact of flavonoids on metabolic disorders.

Author Contributions

M.H.F., S.M.N., and J.K.P. designed the paper. A.K.S., H.K., and G.C.T. collected the literature data. M.H.F., R.K., A.K.P., E.C.-B., and G.A. analyzed the data. G.D., R.G.K., A.K.S., A.K.P., E.C.-B., H.K., and G.C.T. wrote the first draft of the paper, and S.M.N., M.M., and R.B. made further improvements. S.M., C.R.C., and A.B. revised and edited the manuscript and also suggested improvements. All authors read and approved the final manuscript.

Funding

This research received no external funding.

Acknowledgments

A.K.S. and R.K. acknowledge financial support in the form of Senior Research Fellowship from Council of Scientific and Industrial Research, New Delhi, India. A.K.S., R.K. and A.K.P. acknowledge University Grant Commission-Special Assistance Programme and Department of Science and Technology Fund for improvement of science and technology infrastructure and facilities of Biochemistry Department, University of Allahabad.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wellen, K.E.; Hotamisligil, G.S. Inflammation, stress, and diabetes. J. Clin. Investig. 2005, 115, 1111–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. LeRoith, D.; Taylor, S.I.; Olefsky, J.M. Diabetes Mellitus: A Fundamental and Clinical Text; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2004. [Google Scholar]
  3. Hurley, J.V.J.V. Acute Inflammation; Churchill Livingstone: Edinburgh, UK, 1972. [Google Scholar]
  4. Ryan, G.B.; Majno, G. Acute inflammation. A review. Am. J. Pathol. 1977, 86, 183–276. [Google Scholar] [PubMed]
  5. Feghali, C.A.; Wright, T.M. Cytokines in acute and chronic inflammation. Front. Biosci. 1997, 2, 12–26. [Google Scholar]
  6. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
  7. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860. [Google Scholar] [CrossRef] [PubMed]
  8. Hussain, S.P.; Harris, C.C. Inflammation and cancer: An ancient link with novel potentials. Int. J. Cancer 2007, 121, 2373–2380. [Google Scholar] [CrossRef] [PubMed]
  9. Balkwill, F.R.; Mantovani, A. Cancer-related inflammation: Common themes and therapeutic opportunities. Semin. Cancer Biol. 2012, 22, 33–40. [Google Scholar] [CrossRef] [PubMed]
  10. Candido, J.; Hagemann, T. Cancer-related inflammation. J. Clin. Immunol. 2013, 33, S79–S84. [Google Scholar] [CrossRef]
  11. Mathieu, J.; Tissot, M.; Nolibe, D.; Florentin, I.; Kergonou, J.F.; Giroud, J.P. Local and systemic effects of an acute inflammation on eicosanoid generation capacity of polymorphonuclear cells and macrophages. Int. J. Exp. Pathol. 1990, 71, 603–616. [Google Scholar]
  12. Cicchitti, L.; Martelli, M.; Cerritelli, F. Chronic inflammatory disease and osteopathy: A systematic review. PLoS ONE 2015, 10, e0121327. [Google Scholar] [CrossRef]
  13. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
  14. Creely, S.J.; McTernan, P.G.; Kusminski, C.M.; Fisher f, M.; Da Silva, N.F.; Khanolkar, M.; Evans, M.; Harte, A.L.; Kumar, S. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2007, 292, E740–E747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  16. Freigang, S.; Ampenberger, F.; Weiss, A.; Kanneganti, T.-D.; Iwakura, Y.; Hersberger, M.; Kopf, M. Fatty acid–induced mitochondrial uncoupling elicits inflammasome-independent IL-1α and sterile vascular inflammation in atherosclerosis. Nat. Immunol. 2013, 14, 1045. [Google Scholar] [CrossRef] [PubMed]
  17. Libby, P.; Okamoto, Y.; Rocha, V.Z.; Folco, E. Inflammation in atherosclerosis: Transition from theory to practice. Circ. J. 2010, 74, 213–220. [Google Scholar] [CrossRef] [PubMed]
  18. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46. [Google Scholar] [CrossRef] [PubMed]
  19. Naseri, R.; Farzaei, F.; Haratipour, P.; Nabavi, S.F.; Habtemariam, S.; Farzaei, M.H.; Khodarahmi, R.; Tewari, D.; Momtaz, S. Anthocyanins in the Management of Metabolic Syndrome: A Pharmacological and Biopharmaceutical Review. Front. Pharm. 2018, 9. [Google Scholar] [CrossRef]
  20. Farzaei, M.H.; El-Senduny, F.F.; Momtaz, S.; Parvizi, F.; Iranpanah, A.; Tewari, D.; Naseri, R.; Abdolghaffari, A.H.; Rezaei, N. An update on dietary consideration in inflammatory bowel disease: Anthocyanins and more. Expert. Rev. Gastroenterol. Hepatol. 2018, 12, 1007–1024. [Google Scholar] [CrossRef]
  21. Farzaei, M.H.; Bahramsoltani, R.; Abdolghaffari, A.H.; Sodagari, H.R.; Esfahani, S.A.; Rezaei, N. A mechanistic review on plant-derived natural compounds as dietary supplements for prevention of inflammatory bowel disease. Expert. Rev. Gastroenterol. Hepatol. 2016, 10, 745–758. [Google Scholar] [CrossRef]
  22. Liu, K.; Pi, F.; Zhang, H.; Ji, J.; Xia, S.; Cui, F.; Sun, J.; Sun, X. Metabolomics Analysis To Evaluate the Anti-Inflammatory Effects of Polyphenols: Glabridin Reversed Metabolism Change Caused by LPS in RAW 264.7 Cells. J. Agric. Food Chem. 2017, 65, 6070–6079. [Google Scholar] [CrossRef]
  23. Park, S.H.; Kang, J.S.; Yoon, Y.D.; Lee, K.; Kim, K.J.; Lee, K.H.; Lee, C.W.; Moon, E.Y.; Han, S.B.; Kim, B.H. Glabridin inhibits lipopolysaccharide-induced activation of a microglial cell line, BV-2, by blocking NF-κB and AP-1. Phytother. Res. 2010, 24, S29–S34. [Google Scholar] [CrossRef] [PubMed]
  24. Chandrasekaran, C.; Deepak, H.; Thiyagarajan, P.; Kathiresan, S.; Sangli, G.K.; Deepak, M.; Agarwal, A. Dual inhibitory effect of Glycyrrhiza glabra (GutGard™) on COX and LOX products. Phytomedicine 2011, 18, 278–284. [Google Scholar] [CrossRef] [PubMed]
  25. Simmler, C.; Pauli, G.F.; Chen, S.N. Phytochemistry and biological properties of glabridin. Fitoterapia 2013, 90, 160–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Yehuda, I.; Madar, Z.; Leikin-Frenkel, A.; Tamir, S. Glabridin, an isoflavan from licorice root, downregulates iNOS expression and activity under high-glucose stress and inflammation. Mol. Nutr. Food Res. 2015, 59, 1041–1052. [Google Scholar] [CrossRef] [PubMed]
  27. Kim, J.Y.; Kang, J.S.; Kim, H.M.; Ryu, H.S.; Kim, H.S.; Lee, H.K.; Kim, Y.J.; Hong, J.T.; Kim, Y.; Han, S.B. Inhibition of bone marrow-derived dendritic cell maturation by glabridin. Int. Immunopharmacol. 2010, 10, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
  28. Chang, W.-T.; Huang, W.-C.; Liou, C.-J. Evaluation of the anti-inflammatory effects of phloretin and phlorizin in lipopolysaccharide-stimulated mouse macrophages. Food Chem. 2012, 134, 972–979. [Google Scholar] [CrossRef] [PubMed]
  29. Mohamed, S. Functional foods against metabolic syndrome (obesity, diabetes, hypertension and dyslipidemia) and cardiovasular disease. Trends Food Sci. Technol. 2014, 35, 114–128. [Google Scholar] [CrossRef]
  30. Chuang, C.C.; McIntosh, M.K. Potential mechanisms by which polyphenol-rich grapes prevent obesity-mediated inflammation and metabolic diseases. Annu. Rev. Nutr. 2011, 31, 155–176. [Google Scholar] [CrossRef] [PubMed]
  31. Basu, A.; Du, M.; Sanchez, K.; Leyva, M.J.; Betts, N.M.; Blevins, S.; Wu, M.; Aston, C.E.; Lyons, T.J. Green tea minimally affects biomarkers of inflammation in obese subjects with metabolic syndrome. Nutrition 2011, 27, 206–213. [Google Scholar] [CrossRef] [Green Version]
  32. Panahi, Y.; Hosseini, M.S.; Khalili, N.; Naimi, E.; Majeed, M.; Sahebkar, A. Antioxidant and anti-inflammatory effects of curcuminoid-piperine combination in subjects with metabolic syndrome: A randomized controlled trial and an updated meta-analysis. Clin. Nutr. 2015, 34, 1101–1108. [Google Scholar] [CrossRef]
  33. Peluso, I.; Romanelli, L.; Palmery, M. Interactions between prebiotics, probiotics, polyunsaturated fatty acids and polyphenols: Diet or supplementation for metabolic syndrome prevention? Int. J. Food Sci. Nutr. 2014, 65, 259–267. [Google Scholar] [CrossRef] [PubMed]
  34. Amiot, M.J.; Riva, C.; Vinet, A. Effects of dietary polyphenols on metabolic syndrome features in humans: A systematic review. Obes. Rev. 2016, 17, 573–586. [Google Scholar] [CrossRef] [PubMed]
  35. Cherniack, E.P. Polyphenols: Planting the seeds of treatment for the metabolic syndrome. Nutrition 2011, 27, 617–623. [Google Scholar] [CrossRef] [PubMed]
  36. Jungbauer, A.; Medjakovic, S. Anti-inflammatory properties of culinary herbs and spices that ameliorate the effects of metabolic syndrome. Maturitas 2012, 71, 227–239. [Google Scholar] [CrossRef]
  37. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  38. Luo, W.; Morrison, H.; de Groh, M.; Waters, C.; DesMeules, M.; Jones-McLean, E.; Ugnat, A.M.; Desjardins, S.; Lim, M.; Mao, Y. The burden of adult obesity in Canada. Chronic. Dis. Can. 2007, 27, 135–144. [Google Scholar]
  39. Kumar, S.; Pandey, A.K. Free radicals: Health implications and their mitigation by herbals. Br. J. Med. Med. Res. 2015, 7, 438–457. [Google Scholar] [CrossRef]
  40. Sharma, A.K.; Kumar, S.; Chashoo, G.; Saxena, A.K.; Pandey, A.K. Cell cycle inhibitory activity of Piper longum against A549 cell line and its protective effect against metal-induced toxicity in rats. Indian J. Biochem. Biophys. 2014, 51, 358–364. [Google Scholar]
  41. Makowski, L.; Boord, J.B.; Maeda, K.; Babaev, V.R.; Uysal, K.T.; Morgan, M.A.; Parker, R.A.; Suttles, J.; Fazio, S.; Hotamisligil, G.S.; et al. Lack of macrophage fatty-acid-binding protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis. Nat. Med. 2001, 7, 699–705. [Google Scholar] [CrossRef]
  42. Bouloumie, A.; Sengenes, C.; Portolan, G.; Galitzky, J.; Lafontan, M. Adipocyte produces matrix metalloproteinases 2 and 9: Involvement in adipose differentiation. Diabetes 2001, 50, 2080–2086. [Google Scholar] [CrossRef]
  43. Mantovani, A.; Sica, A.; Locati, M. Macrophage polarization comes of age. Immunity 2005, 23, 344–346. [Google Scholar] [CrossRef] [PubMed]
  44. Laskin, D.L. Macrophages and inflammatory mediators in chemical toxicity: A battle of forces. Chem. Res. Toxicol 2009, 22, 1376–1385. [Google Scholar] [CrossRef] [PubMed]
  45. Baker, R.G.; Hayden, M.S.; Ghosh, S. NF-kappaB, inflammation, and metabolic disease. Cell Metab. 2011, 13, 11–22. [Google Scholar] [CrossRef] [PubMed]
  46. Tenoever, B.R.; Ng, S.L.; Chua, M.A.; McWhirter, S.M.; Garcia-Sastre, A.; Maniatis, T. Multiple functions of the IKK-related kinase IKKepsilon in interferon-mediated antiviral immunity. Science 2007, 315, 1274–1278. [Google Scholar] [CrossRef] [PubMed]
  47. Chiang, S.H.; Bazuine, M.; Lumeng, C.N.; Geletka, L.M.; Mowers, J.; White, N.M.; Ma, J.T.; Zhou, J.; Qi, N.; Westcott, D.; et al. The protein kinase IKKepsilon regulates energy balance in obese mice. Cell 2009, 138, 961–975. [Google Scholar] [CrossRef] [PubMed]
  48. Weisberg, S.P.; Hunter, D.; Huber, R.; Lemieux, J.; Slaymaker, S.; Vaddi, K.; Charo, I.; Leibel, R.L.; Ferrante, A.W., Jr. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J. Clin. Investig. 2006, 116, 115–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Rocha, V.Z.; Libby, P. Obesity, inflammation, and atherosclerosis. Nat. Rev. Cardiol. 2009, 6, 399–409. [Google Scholar] [CrossRef] [PubMed]
  50. Kumar, S.; Kumar, R.; Dwivedi, A.; Pandey, A.K. In vitro antioxidant, antibacterial, and cytotoxic activity and in vivo effect of Syngonium podophyllum and Eichhornia crassipes leaf extracts on isoniazid induced oxidative stress and hepatic markers. Biomed. Res. Int. 2014, 2014. [Google Scholar] [CrossRef]
  51. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013. [Google Scholar] [CrossRef]
  52. Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
  53. Lin, Z.; Kumar, A.; SenBanerjee, S.; Staniszewski, K.; Parmar, K.; Vaughan, D.E.; Gimbrone Jr, M.A.; Balasubramanian, V.; García-Cardeña, G.; Jain, M.K. Kruppel-like factor 2 (KLF2) regulates endothelial thrombotic function. Circ. Res. 2005, 96, 48–57. [Google Scholar] [CrossRef] [PubMed]
  54. Houstis, N.; Rosen, E.D.; Lander, E.S. Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 2006, 440, 944. [Google Scholar] [CrossRef] [PubMed]
  55. Chrysant, S. A new paradigm in the treatment of the cardiovascular disease continuum: Focus on prevention. Hippokratia 2011, 15, 7. [Google Scholar] [PubMed]
  56. Awan, Z.; Genest, J. Inflammation modulation and cardiovascular disease prevention. Eur. J. Prev. Cardiol. 2014, 22, 719–733. [Google Scholar] [CrossRef] [PubMed]
  57. Ruparelia, N.; Chai, J.T.; Fisher, E.A.; Choudhury, R.P. Inflammatory processes in cardiovascular disease: A route to targeted therapies. Nat. Rev. Cardiol 2017, 14. [Google Scholar] [CrossRef] [PubMed]
  58. Jia, H.; Abtahian, F.; Aguirre, A.D.; Lee, S.; Chia, S.; Lowe, H.; Kato, K.; Yonetsu, T.; Vergallo, R.; Hu, S.; et al. In vivo diagnosis of plaque erosion and calcified nodule in patients with acute coronary syndrome by intravascular optical coherence tomography. J. Am. Coll. Cardiol. 2013, 62, 1748–1758. [Google Scholar] [CrossRef] [PubMed]
  59. Frangogiannis, N.G. Regulation of the inflammatory response in cardiac repair. Circ. Res. 2012, 110, 159–173. [Google Scholar] [CrossRef] [PubMed]
  60. Prondzinsky, R.; Unverzagt, S.; Lemm, H.; Wegener, N.A.; Schlitt, A.; Heinroth, K.M.; Dietz, S.; Buerke, U.; Kellner, P.; Loppnow, H.; et al. Interleukin-6, -7, -8 and -10 predict outcome in acute myocardial infarction complicated by cardiogenic shock. Clin. Res. Cardiol. 2012, 101, 375–384. [Google Scholar] [CrossRef]
  61. Vinten-Johansen, J. Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovasc. Res. 2004, 61, 481–497. [Google Scholar] [CrossRef] [Green Version]
  62. Nahrendorf, M.; Swirski, F.K.; Aikawa, E.; Stangenberg, L.; Wurdinger, T.; Figueiredo, J.L.; Libby, P.; Weissleder, R.; Pittet, M.J. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J. Exp. Med. 2007, 204, 3037–3047. [Google Scholar] [CrossRef] [Green Version]
  63. Nourshargh, S.; Renshaw, S.A.; Imhof, B.A. Reverse Migration of Neutrophils: Where, When, How, and Why? Trends Immunol. 2016, 37, 273–286. [Google Scholar] [CrossRef] [PubMed]
  64. Oyama, J.; Blais, C., Jr.; Liu, X.; Pu, M.; Kobzik, L.; Kelly, R.A.; Bourcier, T. Reduced myocardial ischemia-reperfusion injury in toll-like receptor 4-deficient mice. Circulation 2004, 109, 784–789. [Google Scholar] [CrossRef] [PubMed]
  65. Riad, A.; Jager, S.; Sobirey, M.; Escher, F.; Yaulema-Riss, A.; Westermann, D.; Karatas, A.; Heimesaat, M.M.; Bereswill, S.; Dragun, D.; et al. Toll-like receptor-4 modulates survival by induction of left ventricular remodeling after myocardial infarction in mice. J. Immunol. 2008, 180, 6954–6961. [Google Scholar] [CrossRef] [PubMed]
  66. Ruparelia, N.; Godec, J.; Lee, R.; Chai, J.T.; Dall’Armellina, E.; McAndrew, D.; Digby, J.E.; Forfar, J.C.; Prendergast, B.D.; Kharbanda, R.K.; et al. Acute myocardial infarction activates distinct inflammation and proliferation pathways in circulating monocytes, prior to recruitment, and identified through conserved transcriptional responses in mice and humans. Eur. Heart J. 2015, 36, 1923–1934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Takahashi, M. NLRP3 inflammasome as a novel player in myocardial infarction. Int. Heart J. 2014, 55, 101–105. [Google Scholar] [CrossRef] [PubMed]
  68. Distelmaier, K.; Adlbrecht, C.; Jakowitsch, J.; Winkler, S.; Dunkler, D.; Gerner, C.; Wagner, O.; Lang, I.M.; Kubicek, M. Local complement activation triggers neutrophil recruitment to the site of thrombus formation in acute myocardial infarction. Thromb. Haemost. 2009, 102, 564–572. [Google Scholar] [CrossRef] [PubMed]
  69. Sellak, H.; Franzini, E.; Hakim, J.; Pasquier, C. Reactive oxygen species rapidly increase endothelial ICAM-1 ability to bind neutrophils without detectable upregulation. Blood 1994, 83, 2669–2677. [Google Scholar] [Green Version]
  70. Libby, P. Inflammation and cardiovascular disease mechanisms. Am. J. Clin. Nutr 2006, 83, 456–460. [Google Scholar] [CrossRef]
  71. Libby, P.; Ridker, P.M.; Maseri, A. Inflammation and atherosclerosis. Circulation 2002, 105, 1135–1143. [Google Scholar] [CrossRef]
  72. Pearson, T.A.; Mensah, G.A.; Alexander, R.W.; Anderson, J.L.; Cannon, R.O., 3rd; Criqui, M.; Fadl, Y.Y.; Fortmann, S.P.; Hong, Y.; Myers, G.L.; et al. Markers of inflammation and cardiovascular disease: Application to clinical and public health practice: A statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation 2003, 107, 499–511. [Google Scholar] [CrossRef]
  73. Golia, E.; Limongelli, G.; Natale, F.; Fimiani, F.; Maddaloni, V.; Pariggiano, I.; Bianchi, R.; Crisci, M.; D’Acierno, L.; Giordano, R.; et al. Inflammation and cardiovascular disease: From pathogenesis to therapeutic target. Curr. Atheroscler Rep. 2014, 16. [Google Scholar] [CrossRef] [PubMed]
  74. Cybulsky, M.I.; Iiyama, K.; Li, H.; Zhu, S.; Chen, M.; Iiyama, M.; Davis, V.; Gutierrez-Ramos, J.-C.; Connelly, P.W.; Milstone, D.S. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J. Clin. Investig. 2001, 107, 1255–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Sever, P.S.; Dahlöf, B.; Poulter, N.R.; Wedel, H.; Beevers, G.; Caulfield, M.; Collins, R.; Kjeldsen, S.E.; Kristinsson, A.; McInnes, G.T. Prevention of coronary and stroke events with atorvastatin in hypertensive patients who have average or lower-than-average cholesterol concentrations, in the Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering Arm (ASCOT-LLA): A multicentre randomised controlled trial. Lancet 2003, 361, 1149–1158. [Google Scholar] [PubMed]
  76. Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte chemoattractant protein-1 (MCP-1): An overview. J. Interferon Cytokine Res. 2009, 29, 313–326. [Google Scholar] [CrossRef] [PubMed]
  77. Khovidhunkit, W.; Kim, M.S.; Memon, R.A.; Shigenaga, J.K.; Moser, A.H.; Feingold, K.R.; Grunfeld, C. Effects of infection and inflammation on lipid and lipoprotein metabolism: Mechanisms and consequences to the host. J. Lipid Res. 2004, 45, 1169–1196. [Google Scholar] [CrossRef]
  78. Feingold, K.R.; Grunfeld, C. The acute phase response inhibits reverse cholesterol transport. J. Lipid Res. 2010, 51, 682–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Annema, W.; Nijstad, N.; Tolle, M.; de Boer, J.F.; Buijs, R.V.; Heeringa, P.; van der Giet, M.; Tietge, U.J. Myeloperoxidase and serum amyloid A contribute to impaired in vivo reverse cholesterol transport during the acute phase response but not group IIA secretory phospholipase A(2). J. Lipid Res. 2010, 51, 743–754. [Google Scholar] [CrossRef] [PubMed]
  80. Ronda, M.C.; Dijkhorst-Oei, L.T.; Rutten, G.E. Reasons and barriers for using a patient portal: Survey among patients with diabetes mellitus. J. Med. Internet Res. 2014, 16. [Google Scholar] [CrossRef] [PubMed]
  81. Tabas, I.; Williams, K.J.; Boren, J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: Update and therapeutic implications. Circulation 2007, 116, 1832–1844. [Google Scholar] [CrossRef]
  82. Cybulsky, M.I.; Gimbrone, M.A., Jr. Endothelial expression of a mononuclear leukocyte adhesion molecule during atherogenesis. Science 1991, 251, 788–791. [Google Scholar] [CrossRef]
  83. Shao, B.Z.; Han, B.Z.; Zeng, Y.X.; Su, D.F.; Liu, C. The roles of macrophage autophagy in atherosclerosis. Acta Pharm. Sin. 2016, 37, 150–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Gharavi, N.M.; Alva, J.A.; Mouillesseaux, K.P.; Lai, C.; Yeh, M.; Yeung, W.; Johnson, J.; Szeto, W.L.; Hong, L.; Fishbein, M.; et al. Role of the Jak/STAT pathway in the regulation of interleukin-8 transcription by oxidized phospholipids in vitro and in atherosclerosis in vivo. J. Biol. Chem. 2007, 282, 31460–31468. [Google Scholar] [CrossRef] [PubMed]
  85. Hellings, W.E.; Peeters, W.; Moll, F.L.; Piers, S.R.; van Setten, J.; Van der Spek, P.J.; de Vries, J.-P.P.; Seldenrijk, K.A.; De Bruin, P.C.; Vink, A. Composition of carotid atherosclerotic plaque is associated with cardiovascular outcome: A prognostic study. Circulation 2010, 121, 1941–1950. [Google Scholar] [CrossRef] [PubMed]
  86. Vink, A.; Schoneveld, A.H.; Lamers, D.; Houben, A.J.; van der Groep, P.; van Diest, P.J.; Pasterkamp, G. HIF-1 alpha expression is associated with an atheromatous inflammatory plaque phenotype and upregulated in activated macrophages. Atherosclerosis 2007, 195, 69–75. [Google Scholar] [CrossRef] [PubMed]
  87. Folco, E.J.; Sheikine, Y.; Rocha, V.Z.; Christen, T.; Shvartz, E.; Sukhova, G.K.; Di Carli, M.F.; Libby, P. Hypoxia but not inflammation augments glucose uptake in human macrophages: Implications for imaging atherosclerosis with 18fluorine-labeled 2-deoxy-D-glucose positron emission tomography. J. Am. Coll. Cardiol. 2011, 58, 603–614. [Google Scholar] [CrossRef] [PubMed]
  88. Wild, S.; Roglic, G.; Green, A.; Sicree, R.; King, H. Global prevalence of diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care 2004, 27, 1047–1053. [Google Scholar] [CrossRef] [PubMed]
  89. Semenkovich, C.F. Insulin resistance and atherosclerosis. J. Clin. Investig. 2006, 116, 1813–1822. [Google Scholar] [CrossRef] [PubMed]
  90. King, A.J. The use of animal models in diabetes research. Br. J. Pharmacol. 2012, 166, 877–894. [Google Scholar] [CrossRef] [Green Version]
  91. Singh, R.; Shaw, J.; Zimmet, P. Epidemiology of childhood type 2 diabetes in the developing world. Pediatr Diabetes 2004, 5, 154–168. [Google Scholar] [CrossRef]
  92. Atkinson, M.A.; Eisenbarth, G.S.; Michels, A.W. Type 1 diabetes. Lancet 2014, 383, 69–82. [Google Scholar] [CrossRef]
  93. Atkinson, M.A. The pathogenesis and natural history of type 1 diabetes. Cold Spring Harb. Perspect. Med. 2012, 2. [Google Scholar] [CrossRef] [PubMed]
  94. Yoon, J.W.; Jun, H.S. Autoimmune destruction of pancreatic beta cells. Am. J. 2005, 12, 580–591. [Google Scholar]
  95. van Lummel, M.; Zaldumbide, A.; Roep, B.O. Changing faces, unmasking the beta-cell: Post-translational modification of antigens in type 1 diabetes. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 299–306. [Google Scholar] [CrossRef] [PubMed]
  96. Vcelakova, J.; Blatny, R.; Halbhuber, Z.; Kolar, M.; Neuwirth, A.; Petruzelkova, L.; Ulmannova, T.; Kolouskova, S.; Sumnik, Z.; Pithova, P. The effect of diabetes-associated autoantigens on cell processes in human PBMCs and their relevance to autoimmune diabetes development. J. Diabetes Res. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  97. Mauvais, F.X.; Diana, J.; van Endert, P. Beta cell antigens in type 1 diabetes: Triggers in pathogenesis and therapeutic targets. F1000Research 2016, 5. [Google Scholar] [CrossRef] [PubMed]
  98. Zhao, Y.; Krishnamurthy, B.; Mollah, Z.U.; Kay, T.W.; Thomas, H.E. NF-kappaB in type 1 diabetes. Inflamm. Allergy Drug Targets 2011, 10, 208–217. [Google Scholar] [CrossRef]
  99. Limbert, C. Type 1 diabetes-an auto-inflammatory disease: A new concept, new therapeutical strategies. J. Transl. Med. 2012, 10, I12. [Google Scholar] [CrossRef]
  100. Hohmeier, H.E.; Tran, V.V.; Chen, G.; Gasa, R.; Newgard, C.B. Inflammatory mechanisms in diabetes: Lessons from the beta-cell. Int J. Obes Relat Metab Disord 2003, 27, 12–16. [Google Scholar] [CrossRef]
  101. Kaneto, H.; Matsuoka, T.A.; Katakami, N.; Kawamori, D.; Miyatsuka, T.; Yoshiuchi, K.; Yasuda, T.; Sakamoto, K.; Yamasaki, Y.; Matsuhisa, M. Oxidative stress and the JNK pathway are involved in the development of type 1 and type 2 diabetes. Curr. Mol. Med. 2007, 7, 674–686. [Google Scholar] [CrossRef]
  102. Nishiyama, T.; Mae, T.; Kishida, H.; Tsukagawa, M.; Mimaki, Y.; Kuroda, M.; Sashida, Y.; Takahashi, K.; Kawada, T.; Nakagawa, K.; et al. Curcuminoids and sesquiterpenoids in turmeric (Curcuma longa L.) suppress an increase in blood glucose level in type 2 diabetic KK-Ay mice. J. Agric. Food Chem. 2005, 53, 959–963. [Google Scholar] [CrossRef]
  103. Chawla, A.; Nguyen, K.D.; Goh, Y.P. Macrophage-mediated inflammation in metabolic disease. Nat. Rev. Immunol. 2011, 11, 738–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Mavrogiannaki, A.N.; Migdalis, I.N. Nonalcoholic Fatty liver disease, diabetes mellitus and cardiovascular disease: Newer data. Int. J. Endocrinol. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  105. Saltiel, A.R.; Pessin, J.E. Insulin signaling pathways in time and space. Trends Cell Biol. 2002, 12, 65–71. [Google Scholar] [CrossRef]
  106. Rehman, K.; Akash, M.S. Mechanisms of inflammatory responses and development of insulin resistance: How are they interlinked? J. Biomed. Sci. 2016, 23. [Google Scholar] [CrossRef] [PubMed]
  107. Badawi, A.; Klip, A.; Haddad, P.; Cole, D.E.; Bailo, B.G.; El-Sohemy, A.; Karmali, M. Type 2 diabetes mellitus and inflammation: Prospects for biomarkers of risk and nutritional intervention. Diabetes Metab. Syndr. Obes. 2010, 3, 173–186. [Google Scholar] [CrossRef] [PubMed]
  108. Serrano-Marco, L.; Barroso, E.; El Kochairi, I.; Palomer, X.; Michalik, L.; Wahli, W.; Vazquez-Carrera, M. The peroxisome proliferator-activated receptor (PPAR) beta/delta agonist GW501516 inhibits IL-6-induced signal transducer and activator of transcription 3 (STAT3) activation and insulin resistance in human liver cells. Diabetologia 2012, 55, 743–751. [Google Scholar] [CrossRef] [PubMed]
  109. Lukic, L.; Lalic, N.M.; Rajkovic, N.; Jotic, A.; Lalic, K.; Milicic, T.; Seferovic, J.P.; Macesic, M.; Gajovic, J.S. Hypertension in obese type 2 diabetes patients is associated with increases in insulin resistance and IL-6 cytokine levels: Potential targets for an efficient preventive intervention. Int. J. Env. Res. Public. Health 2014, 11, 3586–3598. [Google Scholar] [CrossRef] [PubMed]
  110. Kawaguchi, T.; Shiba, N.; Takano, Y.; Maeda, T.; Sata, M. Hybrid training of voluntary and electrical muscle contractions decreased fasting blood glucose and serum interleukin-6 levels in elderly people: A pilot study. Appl. Physiol. Nutr. Metab. 2011, 36, 276–283. [Google Scholar] [CrossRef] [PubMed]
  111. Dou, L.; Zhao, T.; Wang, L.; Huang, X.; Jiao, J.; Gao, D.; Zhang, H.; Shen, T.; Man, Y.; Wang, S.; et al. miR-200s contribute to interleukin-6 (IL-6)-induced insulin resistance in hepatocytes. J. Biol. Chem. 2013, 288, 22596–22606. [Google Scholar] [CrossRef] [PubMed]
  112. Yin, J.; Hao, Z.; Ma, Y.; Liao, S.; Li, X.; Fu, J.; Wu, Y.; Shen, J.; Zhang, P.; Li, X.; et al. Concomitant activation of the PI3K/Akt and ERK1/2 signalling is involved in cyclic compressive force-induced IL-6 secretion in MLO-Y4 cells. Cell Biol. Int. 2014, 38, 591–598. [Google Scholar] [CrossRef] [PubMed]
  113. Kumar, S.; Pandey, A.K. Oxidative Stress-Related MicroRNAs as Diagnostic Markers: A Newer Insight in Diagnostics. In Oxidative Stress: Diagnostic Methods and Applications in Medical Science; Maurya, P.K., Chandra, P., Eds.; Springer: Berlin/Heidelberg, Germany, 2017; pp. 113–125. Available online: https://0-link-springer-com.brum.beds.ac.uk/chapter/10.1007/978-981-10-4711-4_6 (accessed on 29 August 2019).
  114. Kim, T.H.; Choi, S.E.; Ha, E.S.; Jung, J.G.; Han, S.J.; Kim, H.J.; Kim, D.J.; Kang, Y.; Lee, K.W. IL-6 induction of TLR-4 gene expression via STAT3 has an effect on insulin resistance in human skeletal muscle. Acta Diabetol. 2013, 50, 189–200. [Google Scholar] [CrossRef] [PubMed]
  115. Gaggini, M.; Morelli, M.; Buzzigoli, E.; DeFronzo, R.A.; Bugianesi, E.; Gastaldelli, A. Non-alcoholic fatty liver disease (NAFLD) and its connection with insulin resistance, dyslipidemia, atherosclerosis and coronary heart disease. Nutrients 2013, 5, 1544–1560. [Google Scholar] [CrossRef] [PubMed]
  116. Crook, M. Type 2 diabetes mellitus: A disease of the innate immune system? An update. Diabet. Med. 2004, 21, 203–207. [Google Scholar] [CrossRef] [PubMed]
  117. Pickup, J.C. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care 2004, 27, 813–823. [Google Scholar] [CrossRef] [PubMed]
  118. Gregor, M.F.; Hotamisligil, G.S. Inflammatory mechanisms in obesity. Annu. Rev. Immunol. 2011, 29, 415–445. [Google Scholar] [CrossRef] [PubMed]
  119. Klop, B.; Elte, J.W.F.; Cabezas, M.C. Dyslipidemia in obesity: Mechanisms and potential targets. Nutrients 2013, 5, 1218–1240. [Google Scholar] [CrossRef]
  120. Calder, P.C.; Ahluwalia, N.; Brouns, F.; Buetler, T.; Clement, K.; Cunningham, K.; Esposito, K.; Jönsson, L.S.; Kolb, H.; Lansink, M. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br. J. Nutr. 2011, 106, S71–S78. [Google Scholar] [CrossRef] [PubMed]
  121. Bray, G.A.; Frühbeck, G.; Ryan, D.H.; Wilding, J.P.H. Management of obesity. Lancet 2016, 387, 1947–1956. [Google Scholar] [CrossRef] [Green Version]
  122. Li, Z.; Hardij, J.; Bagchi, D.P.; Scheller, E.L.; MacDougald, O.A. Development, regulation, metabolism and function of bone marrow adipose tissues. Bone 2018, 110, 134–140. [Google Scholar] [CrossRef]
  123. Onder, Y.; Green, C.B. Rhythms of metabolism in adipose tissue and mitochondria. Neurobiol. Sleep Circadian Rhythm. 2018, 4, 57–63. [Google Scholar] [CrossRef]
  124. Cawthorn, W.P.; Sethi, J.K. TNF-α and adipocyte biology. Febs. Lett. 2008, 582, 117–131. [Google Scholar] [CrossRef] [PubMed]
  125. McTernan, P.G.; Harte, A.L.; Anderson, L.A.; Green, A.; Smith, S.A.; Holder, J.C.; Barnett, A.H.; Eggo, M.C.; Kumar, S. Insulin and rosiglitazone regulation of lipolysis and lipogenesis in human adipose tissue in vitro. Diabetes 2002, 51, 1493–1498. [Google Scholar] [CrossRef] [PubMed]
  126. de Luca, C.; Olefsky, J.M. Inflammation and insulin resistance. Febs. Lett. 2008, 582, 97–105. [Google Scholar] [CrossRef] [PubMed]
  127. Chen, F.; Castranova, V.; Shi, X.; Demers, L.M. New insights into the role of nuclear factor-κB, a ubiquitous transcription factor in the initiation of diseases. Clin. Chem. 1999, 45, 7–17. [Google Scholar] [PubMed]
  128. Shirane, M.; Hatakeyama, S.; Hattori, K.; Nakayama, K.; Nakayama, K.-I. Common pathway for the ubiquitination of IκBα, IκBβ, and IκBε mediated by the F-box protein FWD1. J. Biol. Chem. 1999, 274, 28169–28174. [Google Scholar] [CrossRef]
  129. DiDonato, J.A.; Hayakawa, M.; Rothwarf, D.M.; Zandi, E.; Karin, M. A cytokine-responsive IκB kinase that activates the transcription factor NF-κB. Nature 1997, 388, 548. [Google Scholar] [CrossRef] [PubMed]
  130. Chen, Z.; Hagler, J.; Palombella, V.J.; Melandri, F.; Scherer, D.; Ballard, D.; Maniatis, T. Signal-induced site-specific phosphorylation targets I kappa B alpha to the ubiquitin-proteasome pathway. Genes. Dev. 1995, 9, 1586–1597. [Google Scholar] [CrossRef]
  131. Israël, A. The IKK complex, a central regulator of NF-κB activation. Cold Spring Harb. Perspect. Biol. 2010, 2. [Google Scholar] [CrossRef]
  132. Stolarczyk, E. Adipose tissue inflammation in obesity: A metabolic or immune response? Curr. Opin. Pharmacol. 2017, 37, 35–40. [Google Scholar] [CrossRef]
  133. Taleb, S. Inflammation in atherosclerosis. Arch. Cardiovasc. Dis. 2016, 109, 708–715. [Google Scholar] [CrossRef]
  134. Ye, J. Mechanisms of insulin resistance in obesity. Front. Med. 2013, 7, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Heymsfield, S.B.; Wadden, T.A. Mechanisms, Pathophysiology, and Management of Obesity. N. Engl. J. Med. 2017, 376, 254–266. [Google Scholar] [CrossRef] [PubMed]
  136. Benedict, M.; Zhang, X. Non-alcoholic fatty liver disease: An expanded review. World J. Hepatol. 2017, 9, 715–732. [Google Scholar] [CrossRef] [PubMed]
  137. Kawano, Y.; Cohen, D.E. Mechanisms of hepatic triglyceride accumulation in non-alcoholic fatty liver disease. J. Gastroenterol. 2013, 48, 434–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Arrigo, T.; Leonardi, S.; Cuppari, C.; Manti, S.; Lanzafame, A.; D’Angelo, G.; Gitto, E.; Marseglia, L.; Salpietro, C. Role of the diet as a link between oxidative stress and liver diseases. World J. Gastroenterol. 2015, 21, 384. [Google Scholar] [CrossRef] [PubMed]
  139. Niederreiter, L.; Tilg, H. Cytokines and fatty liver diseases. Liver Res. 2018, 2, 14–20. [Google Scholar] [CrossRef]
  140. Singh, A.K.; Bishayee, A.; Pandey, A.K. Targeting Histone Deacetylases with Natural and Synthetic Agents: An Emerging Anticancer Strategy. Nutrients 2018, 10, 731. [Google Scholar] [CrossRef] [PubMed]
  141. Kubes, P.; Mehal, W.Z. Sterile inflammation in the liver. Gastroenterology 2012, 143, 1158–1172. [Google Scholar] [CrossRef] [PubMed]
  142. Tilg, H. Cytokines and liver diseases. Can. J. Gastroenterol. 2001, 15, 661–668. [Google Scholar] [CrossRef] [PubMed]
  143. Tilg, H.; Moschen, A.R. Inflammatory mechanisms in the regulation of insulin resistance. Mol. Med. 2008, 14, 222–231. [Google Scholar] [CrossRef] [PubMed]
  144. Utzschneider, K.M.; Kahn, S.E. Review: The role of insulin resistance in nonalcoholic fatty liver disease. J. Clin. Endocrinol. Metab. 2006, 91, 4753–4761. [Google Scholar] [CrossRef] [PubMed]
  145. Tilg, H.; Moschen, A.R. Evolution of inflammation in nonalcoholic fatty liver disease: The multiple parallel hits hypothesis. Hepatology 2010, 52, 1836–1846. [Google Scholar] [CrossRef] [PubMed]
  146. Gentile, C.L.; Frye, M.A.; Pagliassotti, M.J. Fatty acids and the endoplasmic reticulum in nonalcoholic fatty liver disease. Biofactors 2011, 37, 8–16. [Google Scholar] [CrossRef] [PubMed]
  147. Cerpa-Cruz, S.; González-Díaz, V.; Martínez-Bonilla, G.; Gutiérrez-Ureña, S.; Rodríguez-Cortés, E. Non-Alcoholic Fatty Steatohepatitis an Inflammatory Disorder Beyond the Liver. J. Clin. Cell. Immunol. 2013, 4, 2. [Google Scholar] [CrossRef]
  148. Seki, E.; Brenner, D.A.; Karin, M. A liver full of JNK: Signaling in regulation of cell function and disease pathogenesis, and clinical approaches. Gastroenterology 2012, 143, 307–320. [Google Scholar] [CrossRef] [PubMed]
  149. Tarantino, G.; Caputi, A. JNKs, insulin resistance and inflammation: A possible link between NAFLD and coronary artery disease. World J. Gastroenterol. 2011, 17, 3785. [Google Scholar] [CrossRef] [PubMed]
  150. Zeng, L.; Tang, W.J.; Yin, J.J.; Zhou, B.J. Signal transductions and nonalcoholic fatty liver: A mini-review. Int. J. Clin. Exp. Med. 2014, 7, 1624–1631. [Google Scholar] [PubMed]
  151. McClain, C.J.; Cohen, D.A.; Dinarello, C.A.; Cannon, J.G.; Shedlofsky, S.I.; Kaplan, A.M. Serum interleukin-1 (IL-1) activity in alcoholic hepatitis. Life Sci. 1986, 39, 1479–1485. [Google Scholar] [CrossRef]
  152. Bird, G.L.; Sheron, N.; Goka, A.K.; Alexander, G.J.; Williams, R.S. Increased plasma tumor necrosis factor in severe alcoholic hepatitis. Ann. Intern. Med. 1990, 112, 917–920. [Google Scholar] [CrossRef] [PubMed]
  153. Dominguez, M.; Miquel, R.; Colmenero, J.; Moreno, M.; Garcia-Pagan, J.C.; Bosch, J.; Arroyo, V.; Gines, P.; Caballeria, J.; Bataller, R. Hepatic expression of CXC chemokines predicts portal hypertension and survival in patients with alcoholic hepatitis. Gastroenterology 2009, 136, 1639–1650. [Google Scholar] [CrossRef]
  154. Spahr, L.; Garcia, I.; Bresson-Hadni, S.; Rubbia-Brandt, L.; Guler, R.; Olleros, M.; Chvatchko, Y.; Hadengue, A. Circulating concentrations of interleukin-18, interleukin-18 binding protein, and gamma interferon in patients with alcoholic hepatitis. Liver Int. 2004, 24, 582–587. [Google Scholar] [CrossRef] [PubMed]
  155. Lemmers, A.; Moreno, C.; Gustot, T.; Marechal, R.; Degre, D.; Demetter, P.; de Nadai, P.; Geerts, A.; Quertinmont, E.; Vercruysse, V.; et al. The interleukin-17 pathway is involved in human alcoholic liver disease. Hepatology 2009, 49, 646–657. [Google Scholar] [CrossRef] [PubMed]
  156. Ludwig, J.; Viggiano, T.R.; McGill, D.B.; Oh, B.J. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo. Clin. Proc. 1980, 55, 434–438. [Google Scholar]
  157. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Diehl, A.M.; Brunt, E.M.; Cusi, K.; Charlton, M.; Sanyal, A.J. The diagnosis and management of non-alcoholic fatty liver disease: Practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology 2012, 55, 2005–2023. [Google Scholar] [CrossRef] [PubMed]
  158. Del Campo, J.A.; Gallego, P.; Grande, L. Role of inflammatory response in liver diseases: Therapeutic strategies. World J. Hepatol 2018, 10, 1–7. [Google Scholar] [CrossRef] [PubMed]
  159. Brunt, E.M.; Wong, V.W.; Nobili, V.; Day, C.P.; Sookoian, S.; Maher, J.J.; Bugianesi, E.; Sirlin, C.B.; Neuschwander-Tetri, B.A.; Rinella, M.E. Nonalcoholic fatty liver disease. Nat. Rev. Dis Primers 2015, 1. [Google Scholar] [CrossRef]
  160. Hassan, K.; Bhalla, V.; El Regal, M.E.; A-Kader, H.H. Nonalcoholic fatty liver disease: A comprehensive review of a growing epidemic. World J. Gastroenterol. 2014, 20, 12082–12101. [Google Scholar] [CrossRef] [PubMed]
  161. Chisholm, J.W.; Hong, J.; Mills, S.A.; Lawn, R.M. The LXR ligand T0901317 induces severe lipogenesis in the db/db diabetic mouse. J. Lipid Res. 2003, 44, 2039–2048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Diehl, A.M.; Li, Z.P.; Lin, H.Z.; Yang, S.Q. Cytokines and the pathogenesis of non-alcoholic steatohepatitis. Gut 2005, 54, 303–306. [Google Scholar] [CrossRef] [Green Version]
  163. Wree, A.; Eguchi, A.; McGeough, M.D.; Pena, C.A.; Johnson, C.D.; Canbay, A.; Hoffman, H.M.; Feldstein, A.E. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014, 59, 898–910. [Google Scholar] [CrossRef]
  164. Tilg, H.; Diehl, A.M. Cytokines in alcoholic and nonalcoholic steatohepatitis. N. Engl. J. Med. 2000, 343, 1467–1476. [Google Scholar] [CrossRef] [PubMed]
  165. Martinon, F.; Burns, K.; Tschopp, J. The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  166. Ozaki, E.; Campbell, M.; Doyle, S.L. Targeting the NLRP3 inflammasome in chronic inflammatory diseases: Current perspectives. J. Inflamm. Res. 2015, 8, 15–27. [Google Scholar] [CrossRef] [PubMed]
  167. Sarasota, F. Global Flavonoids Market will Reach USD 1047.63 million in 2021: Zion Market Research. Available online: http://www.econotimes.com/Global-FlavonoidsMarket-will-reach-USD-104763-million-in-2021-ZionMarket-Research-457514 (accessed on 29 August 2019).
  168. Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  169. Crozier, A.; Burns, J.; Aziz, A.A.; Stewart, A.J.; Rabiasz, H.S.; Jenkins, G.I.; Edwards, C.A.; Lean, M.E. Antioxidant flavonols from fruits, vegetables and beverages: Measurements and bioavailability. Biol. Res. 2000, 33, 79–88. [Google Scholar] [CrossRef] [PubMed]
  170. Serafini, M.; Peluso, I.; Raguzzini, A. Flavonoids as anti-inflammatory agents. Proc. Nutr. Soc. 2010, 69, 273–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Vuolo, M.M.; Lima, V.S.; Junior, M.R.M. Phenolic Compounds: Structure, Classification, and Antioxidant Power. In Bioactive Compounds; Segura Campos, M.R., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 33–50. Available online: https://0-www-elsevier-com.brum.beds.ac.uk/books/bioactive-compounds/campos/978-0-12-814774-0 (accessed on 29 August 2019).
  172. Hollman, P.C.; Katan, M.B. Dietary flavonoids: Intake, health effects and bioavailability. Food Chem. Toxicol. 1999, 37, 937–942. [Google Scholar] [CrossRef]
  173. van Dam, R.M.; Naidoo, N.; Landberg, R. Dietary flavonoids and the development of type 2 diabetes and cardiovascular diseases: Review of recent findings. Curr. Opin. Lipidol. 2013, 24, 25–33. [Google Scholar] [CrossRef] [PubMed]
  174. Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet: A Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef] [PubMed]
  175. Pham-Huy, L.A.; He, H.; Pham-Huy, C. Free radicals, antioxidants in disease and health. Int. J. Biomed. Sci. 2008, 4, 89–96. [Google Scholar] [PubMed]
  176. Suen, J.; Thomas, J.; Kranz, A.; Vun, S.; Miller, M. Effect of Flavonoids on Oxidative Stress and Inflammation in Adults at Risk of Cardiovascular Disease: A Systematic Review. Healthcare 2016, 4, 69. [Google Scholar] [CrossRef] [PubMed]
  177. Kim, H.P.; Son, K.H.; Chang, H.W.; Kang, S.S. Anti-inflammatory plant flavonoids and cellular action mechanisms. J. Pharmacol. Sci. 2004, 96, 229–245. [Google Scholar] [CrossRef] [PubMed]
  178. Kimata, M.; Shichijo, M.; Miura, T.; Serizawa, I.; Inagaki, N.; Nagai, H. Effects of luteolin, quercetin and baicalein on immunoglobulin E-mediated mediator release from human cultured mast cells. Clin. Exp. Allergy 2000, 30, 501–508. [Google Scholar] [CrossRef] [PubMed]
  179. Manthey, J.A.; Grohmann, K.; Montanari, A.; Ash, K.; Manthey, C.L. Polymethoxylated flavones derived from citrus suppress tumor necrosis factor-alpha expression by human monocytes. J. Nat. Prod. 1999, 62, 441–444. [Google Scholar] [CrossRef] [PubMed]
  180. Devi, K.P.; Kiruthiga, P.V.; Pandian, S.K. Emerging Role of Flavonoids in Inhibition of NF-κB-Mediated Signaling Pathway: A Review. Int. J. Biomed. Pharm. Sci. 2009, 3, 31–45. [Google Scholar]
  181. Das, H.; Kumar, A.; Lin, Z.; Patino, W.D.; Hwang, P.M.; Feinberg, M.W.; Majumder, P.K.; Jain, M.K. Kruppel-like factor 2 (KLF2) regulates proinflammatory activation of monocytes. Proc. Natl. Acad. Sci. USA 2006, 103, 6653–6658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Schonthaler, H.B.; Guinea-Viniegra, J.; Wagner, E.F. Targeting inflammation by modulating the Jun/AP-1 pathway. Ann. Rheum. Dis 2011, 70, 109–112. [Google Scholar] [CrossRef] [PubMed]
  183. Beretz, A.; Cazenave, J.P. The effect of flavonoids on blood-vessel wall interactions. Prog. Clin. Biol. Res. 1988, 280, 187–200. [Google Scholar] [PubMed]
  184. Geraets, L.; Moonen, H.J.; Brauers, K.; Wouters, E.F.; Bast, A.; Hageman, G.J. Dietary flavones and flavonoles are inhibitors of poly(ADP-ribose)polymerase-1 in pulmonary epithelial cells. J. Nutr. 2007, 137, 2190–2195. [Google Scholar] [CrossRef] [PubMed]
  185. Geraets, L.; Moonen, H.J.; Brauers, K.; Gottschalk, R.W.; Wouters, E.F.; Bast, A.; Hageman, G.J. Flavone as PARP-1 inhibitor: Its effect on lipopolysaccharide induced gene-expression. Eur. J. Pharmacol. 2007, 573, 241–248. [Google Scholar] [CrossRef] [PubMed]
  186. Nakajima, H.; Nagaso, H.; Kakui, N.; Ishikawa, M.; Hiranuma, T.; Hoshiko, S. Critical role of the automodification of poly(ADP-ribose) polymerase-1 in nuclear factor-kappaB-dependent gene expression in primary cultured mouse glial cells. J. Biol. Chem. 2004, 279, 42774–42786. [Google Scholar] [CrossRef] [PubMed]
  187. Esposito, D.; Chen, A.; Grace, M.H.; Komarnytsky, S.; Lila, M.A. Inhibitory effects of wild blueberry anthocyanins and other flavonoids on biomarkers of acute and chronic inflammation in vitro. J. Agric. Food Chem. 2014, 62, 7022–7028. [Google Scholar] [CrossRef] [PubMed]
  188. Mao, Z.; Gan, C.; Zhu, J.; Ma, N.; Wu, L.; Wang, L.; Wang, X. Anti-atherosclerotic activities of flavonoids from the flowers of Helichrysum arenarium L. MOENCH through the pathway of anti-inflammation. Bioorg. Med. Chem. Lett. 2017, 27, 2812–2817. [Google Scholar] [CrossRef] [PubMed]
  189. Garcia-Lafuente, A.; Guillamon, E.; Villares, A.; Rostagno, M.A.; Martinez, J.A. Flavonoids as anti-inflammatory agents: Implications in cancer and cardiovascular disease. Inflamm. Res. 2009, 58, 537–552. [Google Scholar] [CrossRef] [PubMed]
  190. Landberg, R.; Sun, Q.; Rimm, E.B.; Cassidy, A.; Scalbert, A.; Mantzoros, C.S.; Hu, F.B.; van Dam, R.M. Selected dietary flavonoids are associated with markers of inflammation and endothelial dysfunction in U.S. women. J. Nutr. 2011, 141, 618–625. [Google Scholar] [CrossRef]
  191. Steptoe, A.; Gibson, E.L.; Vuononvirta, R.; Hamer, M.; Wardle, J.; Rycroft, J.A.; Martin, J.F.; Erusalimsky, J.D. The effects of chronic tea intake on platelet activation and inflammation: A double-blind placebo controlled trial. Atherosclerosis 2007, 193, 277–282. [Google Scholar] [CrossRef] [PubMed]
  192. Engler, M.B.; Engler, M.M. The emerging role of flavonoid-rich cocoa and chocolate in cardiovascular health and disease. Nutr. Rev. 2006, 64, 109–118. [Google Scholar] [CrossRef]
  193. Wallace, T.C. Anthocyanins in cardiovascular disease. Adv. Nutr 2011, 2, 1–7. [Google Scholar] [CrossRef]
  194. Lotito, S.B.; Frei, B. Dietary flavonoids attenuate tumor necrosis factor alpha-induced adhesion molecule expression in human aortic endothelial cells. Structure-function relationships and activity after first pass metabolism. J. Biol. Chem. 2006, 281, 37102–37110. [Google Scholar] [CrossRef]
  195. Kobuchi, H.; Roy, S.; Sen, C.K.; Nguyen, H.G.; Packer, L. Quercetin inhibits inducible ICAM-1 expression in human endothelial cells through the JNK pathway. Am. J. Physiol. 1999, 277, 403–411. [Google Scholar] [CrossRef]
  196. Sen, C.K.; Bagchi, D. Regulation of inducible adhesion molecule expression in human endothelial cells by grape seed proanthocyanidin extract. Mol. Cell. Biochem. 2001, 216, 1–7. [Google Scholar] [CrossRef] [PubMed]
  197. Chacko, B.K.; Chandler, R.T.; Mundhekar, A.; Khoo, N.; Pruitt, H.M.; Kucik, D.F.; Parks, D.A.; Kevil, C.G.; Barnes, S.; Patel, R.P. Revealing anti-inflammatory mechanisms of soy isoflavones by flow: Modulation of leukocyte-endothelial cell interactions. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, 908–915. [Google Scholar] [CrossRef] [PubMed]
  198. Chacko, B.K.; Chandler, R.T.; D’Alessandro, T.L.; Mundhekar, A.; Khoo, N.K.; Botting, N.; Barnes, S.; Patel, R.P. Anti-inflammatory effects of isoflavones are dependent on flow and human endothelial cell PPARgamma. J. Nutr. 2007, 137, 351–356. [Google Scholar] [CrossRef] [PubMed]
  199. Droke, E.A.; Hager, K.A.; Lerner, M.R.; Lightfoot, S.A.; Stoecker, B.J.; Brackett, D.J.; Smith, B.J. Soy isoflavones avert chronic inflammation-induced bone loss and vascular disease. J. Inflamm. 2007, 4. [Google Scholar] [CrossRef] [PubMed]
  200. Ramiro, E.; Franch, A.; Castellote, C.; Perez-Cano, F.; Permanyer, J.; Izquierdo-Pulido, M.; Castell, M. Flavonoids from Theobroma cacao down-regulate inflammatory mediators. J. Agric. Food Chem. 2005, 53, 8506–8511. [Google Scholar] [CrossRef]
  201. Winterbone, M.S.; Tribolo, S.; Needs, P.W.; Kroon, P.A.; Hughes, D.A. Physiologically relevant metabolites of quercetin have no effect on adhesion molecule or chemokine expression in human vascular smooth muscle cells. Atherosclerosis 2009, 202, 431–438. [Google Scholar] [CrossRef] [PubMed]
  202. Panicker, S.R.; Sreenivas, P.; Babu, M.S.; Karunagaran, D.; Kartha, C.C. Quercetin attenuates monocyte chemoattractant protein-1 gene expression in glucose primed aortic endothelial cells through NF-κB and AP-1. Pharmacol. Res. 2010, 62, 328–336. [Google Scholar] [CrossRef] [PubMed]
  203. Zhu, Y.; Ling, W.; Guo, H.; Song, F.; Ye, Q.; Zou, T.; Li, D.; Zhang, Y.; Li, G.; Xiao, Y.; et al. Anti-inflammatory effect of purified dietary anthocyanin in adults with hypercholesterolemia: A randomized controlled trial. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 843–849. [Google Scholar] [CrossRef]
  204. Risitano, R.; Currò, M.; Cirmi, S.; Ferlazzo, N.; Campiglia, P.; Caccamo, D.; Ientile, R.; Navarra, M. Flavonoid fraction of Bergamot juice reduces LPS-induced inflammatory response through SIRT1-mediated NF-κB inhibition in THP-1 monocytes. PLoS ONE 2014, 9, e107431. [Google Scholar] [CrossRef]
  205. Ku, S.-K.; Bae, J.-S. Baicalin, baicalein and wogonin inhibits high glucose-induced vascular inflammation in vitro and in vivo. BMB Rep. 2015, 48, 519. [Google Scholar] [CrossRef]
  206. Hsueh, T.-P.; Sheen, J.-M.; Pang, J.-H.S.; Bi, K.-W.; Huang, C.-C.; Wu, H.-T.; Huang, S.-T. The anti-atherosclerotic effect of naringin is associated with reduced expressions of cell adhesion molecules and chemokines through NF-κB Pathway. Molecules 2016, 21, 195. [Google Scholar] [CrossRef] [PubMed]
  207. Cirillo, P.; Conte, S.; Cimmino, G.; Pellegrino, G.; Ziviello, F.; Barra, G.; Sasso, F.C.; Borgia, F.; De Palma, R.; Trimarco, B. Nobiletin inhibits oxidized-LDL mediated expression of Tissue Factor in human endothelial cells through inhibition of NF-κB. Biochem. Pharmacol. 2017, 128, 26–33. [Google Scholar] [CrossRef] [PubMed]
  208. Bhaskar, S.; Sudhakaran, P.; Helen, A. Quercetin attenuates atherosclerotic inflammation and adhesion molecule expression by modulating TLR-NF-κB signaling pathway. Cell. Immunol. 2016, 310, 131–140. [Google Scholar] [CrossRef] [PubMed]
  209. Tian, H.; Liu, Q.; Qin, S.; Zong, C.; Zhang, Y.; Yao, S.; Yang, N.; Guan, T.; Guo, S. Synthesis and cardiovascular protective effects of quercetin 7-O-sialic acid. J. Cell. Mol. Med. 2017, 21, 107–120. [Google Scholar] [CrossRef] [PubMed]
  210. Deng, H.-F.; Wang, X.-L.; Sun, H.; Xiao, X.-Z. Puerarin inhibits expression of tissue factor induced by oxidative low-density lipoprotein through activating the PI3K/Akt/eNOS pathway and inhibiting activation of ERK1/2 and NF-κB. Life Sci. 2017, 191, 115–121. [Google Scholar] [CrossRef]
  211. Jeong, Y.J.; Choi, Y.J.; Choi, J.S.; Kwon, H.M.; Kang, S.W.; Bae, J.Y.; Lee, S.S.; Kang, J.S.; Han, S.J.; Kang, Y.H. Attenuation of monocyte adhesion and oxidised LDL uptake in luteolin-treated human endothelial cells exposed to oxidised LDL. Br. J. Nutr. 2007, 97, 447–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Chanet, A.; Milenkovic, D.; Claude, S.; Maier, J.A.; Khan, M.K.; Rakotomanomana, N.; Shinkaruk, S.; Bérard, A.M.; Bennetau-Pelissero, C.; Mazur, A. Flavanone metabolites decrease monocyte adhesion to TNF-α-activated endothelial cells by modulating expression of atherosclerosis-related genes. Br. J. Nutr. 2013, 110, 587–598. [Google Scholar] [CrossRef] [PubMed]
  213. De Andrade, C.; de Sá, M.S.; Toloi, M.T. Effects of phytoestrogens derived from soy bean on expression of adhesion molecules on HUVEC. Climacteric 2012, 15, 186–194. [Google Scholar] [CrossRef] [PubMed]
  214. Lee, S.M.; Lee, Y.J.; Kim, Y.C.; Kim, J.S.; Kang, D.G.; Lee, H.S. Vascular protective role of vitexicarpin isolated from Vitex rotundifolia in human umbilical vein endothelial cells. Inflammation 2012, 35, 584–593. [Google Scholar] [CrossRef] [PubMed]
  215. Xiao, H.-B.; Lu, X.-Y.; Liu, Z.-K.; Luo, Z.-F. Kaempferol inhibits the production of ROS to modulate OPN–α v β 3 integrin pathway in HUVECs. J. Physiol. Biochem. 2016, 72, 303–313. [Google Scholar] [CrossRef] [PubMed]
  216. Lee, I.-C.; Bae, J.-S. Suppressive effects of pelargonidin on PolyPhosphate-mediated vascular inflammatory responses. Arch. Pharmacal. Res. 2017, 40, 258–267. [Google Scholar] [CrossRef] [PubMed]
  217. Shi, J.; Deng, H.; Pan, H.; Xu, Y.; Zhang, M. Epigallocatechin-3-gallate attenuates microcystin-LR induced oxidative stress and inflammation in human umbilical vein endothelial cells. Chemosphere 2017, 168, 25–31. [Google Scholar] [CrossRef] [PubMed]
  218. Ming, X.; Ding, M.; Zhai, B.; Xiao, L.; Piao, T.; Liu, M. Biochanin A inhibits lipopolysaccharide-induced inflammation in human umbilical vein endothelial cells. Life Sci. 2015, 136, 36–41. [Google Scholar] [CrossRef] [PubMed]
  219. Limtrakul, P.; Yodkeeree, S.; Pitchakarn, P.; Punfa, W. Anti-inflammatory effects of proanthocyanidin-rich red rice extract via suppression of MAPK, AP-1 and NF-κB pathways in Raw 264.7 macrophages. Nutr. Res. Pract. 2016, 10, 251–258. [Google Scholar] [CrossRef] [PubMed]
  220. Kunworarath, N.; Rangkadilok, N.; Suriyo, T.; Thiantanawat, A.; Satayavivad, J. Longan (Dimocarpus longan Lour.) inhibits lipopolysaccharide-stimulated nitric oxide production in macrophages by suppressing NF-κB and AP-1 signaling pathways. J. Ethnopharmacol. 2016, 179, 156–161. [Google Scholar] [CrossRef] [PubMed]
  221. Yi, Y.-S.; Cho, J.Y.; Kim, D. Cerbera manghas methanol extract exerts anti-inflammatory activity by targeting c-Jun N-terminal kinase in the AP-1 pathway. J. Ethnopharmacol. 2016, 193, 387–396. [Google Scholar] [CrossRef] [PubMed]
  222. Takada, K.; Seike, T.; Sasaki, T.; Masuda, Y.; Ito, A.; Ishii, H. Nobiletin, a polymethoxyflavone in citrus fruits, reduces TAFI expression in HepG2 cells through transcriptional inhibition. Thromb. Haemost. 2013, 110, 1060–1069. [Google Scholar] [CrossRef] [PubMed]
  223. Lee, S.G.; Kim, B.; Yang, Y.; Pham, T.X.; Park, Y.-K.; Manatou, J.; Koo, S.I.; Chun, O.K.; Lee, J.-Y. Berry anthocyanins suppress the expression and secretion of proinflammatory mediators in macrophages by inhibiting nuclear translocation of NF-κB independent of NRF2-mediated mechanism. J. Nutr. Biochem. 2014, 25, 404–411. [Google Scholar] [CrossRef] [PubMed]
  224. Jun, M.S.; Kim, H.S.; Kim, Y.M.; Kim, H.J.; Park, E.J.; Lee, J.H.; Lee, K.R.; Kim, Y.S.; Chang, K.C. Ethanol Extract of Prunella vulgaris var. lilacina Inhibits HMGB1 Release by Induction of Heme Oxygenase-1 in LPS-activated RAW 264.7 Cells and CLP-induced Septic Mice. Phytother. Res. 2012, 26, 605–612. [Google Scholar] [CrossRef]
  225. Lv, H.; Yu, Z.; Zheng, Y.; Wang, L.; Qin, X.; Cheng, G.; Ci, X. Isovitexin exerts anti-inflammatory and anti-oxidant activities on lipopolysaccharide-induced acute lung injury by inhibiting MAPK and NF-κB and activating HO-1/Nrf2 pathways. Int. J. Biol. Sci. 2016, 12, 72. [Google Scholar] [CrossRef]
  226. Song, Y.S.; Park, C.M. Luteolin and luteolin-7-O-glucoside strengthen antioxidative potential through the modulation of Nrf2/MAPK mediated HO-1 signaling cascade in RAW 264.7 cells. Food Chem. Toxicol. 2014, 65, 70–75. [Google Scholar] [CrossRef] [PubMed]
  227. Cui, X.; Liu, X.; Feng, H.; Zhao, S.; Gao, H. Grape seed proanthocyanidin extracts enhance endothelial nitric oxide synthase expression through 5’-AMP activated protein kinase/Surtuin 1-Krupple like factor 2 pathway and modulate blood pressure in ouabain induced hypertensive rats. Biol. Pharm. Bull. 2012, 35, 2192–2197. [Google Scholar] [CrossRef] [PubMed]
  228. Ceriello, A.; Motz, E. Is oxidative stress the pathogenic mechanism underlying insulin resistance, diabetes, and cardiovascular disease? The common soil hypothesis revisited. Arter. Thromb. Vasc. Biol. 2004, 24, 816–823. [Google Scholar] [CrossRef] [PubMed]
  229. Wollgast, J.; Anklam, E. Review on polyphenols in Theobroma cacao: Changes in composition during the manufacture of chocolate and methodology for identification and quantification. Food Res. Int. 2000, 33, 423–447. [Google Scholar] [CrossRef]
  230. Langer, S.; Marshall, L.J.; Day, A.J.; Morgan, M.R. Flavanols and methylxanthines in commercially available dark chocolate: A study of the correlation with nonfat cocoa solids. J. Agric. Food Chem. 2011, 59, 8435–8441. [Google Scholar] [CrossRef] [PubMed]
  231. Sarria, B.; Martinez-Lopez, S.; Sierra-Cinos, J.L.; Garcia-Diz, L.; Mateos, R.; Bravo, L. Regular consumption of a cocoa product improves the cardiometabolic profile in healthy and moderately hypercholesterolaemic adults. Br. J. Nutr. 2014, 111, 122–134. [Google Scholar] [CrossRef] [PubMed]
  232. Gómez-Juaristi, M.; González-Torres, L.; Bravo, L.; Vaquero, M.; Bastida, S.; Sánchez-Muniz, F. Beneficial effects of chocolate on cardiovascular health. Nutr. Hosp. 2011, 26, 289–292. [Google Scholar] [PubMed]
  233. Monagas, M.; Khan, N.; Andres-Lacueva, C.; Casas, R.; Urpi-Sarda, M.; Llorach, R.; Lamuela-Raventos, R.M.; Estruch, R. Effect of cocoa powder on the modulation of inflammatory biomarkers in patients at high risk of cardiovascular disease. Am. J. Clin. Nutr. 2009, 90, 1144–1150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Wang-Polagruto, J.F.; Villablanca, A.C.; Polagruto, J.A.; Lee, L.; Holt, R.R.; Schrader, H.R.; Ensunsa, J.L.; Steinberg, F.M.; Schmitz, H.H.; Keen, C.L. Chronic consumption of flavanol-rich cocoa improves endothelial function and decreases vascular cell adhesion molecule in hypercholesterolemic postmenopausal women. J. Cardiovasc. Pharm. 2006, 47, 177–186. [Google Scholar] [CrossRef] [PubMed]
  235. Mellor, D.D.; Madden, L.A.; Smith, K.A.; Kilpatrick, E.S.; Atkin, S.L. High-polyphenol chocolate reduces endothelial dysfunction and oxidative stress during acute transient hyperglycaemia in Type 2 diabetes: A pilot randomized controlled trial. Diabet. Med. 2013, 30, 478–483. [Google Scholar] [CrossRef] [PubMed]
  236. Hooper, L.; Kay, C.; Abdelhamid, A.; Kroon, P.A.; Cohn, J.S.; Rimm, E.B.; Cassidy, A. Effects of chocolate, cocoa, and flavan-3-ols on cardiovascular health: A systematic review and meta-analysis of randomized trials. Am. J. Clin. Nutr. 2012, 95, 740–751. [Google Scholar] [CrossRef] [PubMed]
  237. Dower, J.I.; Geleijnse, J.M.; Gijsbers, L.; Zock, P.L.; Kromhout, D.; Hollman, P.C. Effects of the pure flavonoids epicatechin and quercetin on vascular function and cardiometabolic health: A randomized, double-blind, placebo-controlled, crossover trial. Am. J. Clin. Nutr. 2015, 101, 914–921. [Google Scholar] [CrossRef] [PubMed]
  238. Sacanella, E.; Vazquez-Agell, M.; Mena, M.P.; Antunez, E.; Fernandez-Sola, J.; Nicolas, J.M.; Lamuela-Raventos, R.M.; Ros, E.; Estruch, R. Down-regulation of adhesion molecules and other inflammatory biomarkers after moderate wine consumption in healthy women: A randomized trial. Am. J. Clin. Nutr. 2007, 86, 1463–1469. [Google Scholar] [CrossRef]
  239. Kalin, R.; Righi, A.; Del Rosso, A.; Bagchi, D.; Generini, S.; Cerinic, M.M.; Das, D.K. Activin, a grape seed-derived proanthocyanidin extract, reduces plasma levels of oxidative stress and adhesion molecules (ICAM-1, VCAM-1 and E-selectin) in systemic sclerosis. Free Radic. Res. 2002, 36, 819–825. [Google Scholar] [CrossRef] [PubMed]
  240. Karlsen, A.; Paur, I.; Bohn, S.K.; Sakhi, A.K.; Borge, G.I.; Serafini, M.; Erlund, I.; Laake, P.; Tonstad, S.; Blomhoff, R. Bilberry juice modulates plasma concentration of NF-kappaB related inflammatory markers in subjects at increased risk of CVD. Eur. J. Nutr. 2010, 49, 345–355. [Google Scholar] [CrossRef] [PubMed]
  241. Aaby, K.; Skrede, G.; Wrolstad, R.E. Phenolic composition and antioxidant activities in flesh and achenes of strawberries (Fragaria ananassa). J. Agric. Food Chem. 2005, 53, 4032–4040. [Google Scholar] [CrossRef] [PubMed]
  242. Edirisinghe, I.; Banaszewski, K.; Cappozzo, J.; Sandhya, K.; Ellis, C.L.; Tadapaneni, R.; Kappagoda, C.T.; Burton-Freeman, B.M. Strawberry anthocyanin and its association with postprandial inflammation and insulin. Br. J. Nutr. 2011, 106, 913–922. [Google Scholar] [CrossRef]
  243. Ceriello, A.; Esposito, K.; Piconi, L.; Ihnat, M.A.; Thorpe, J.E.; Testa, R.; Boemi, M.; Giugliano, D. Oscillating glucose is more deleterious to endothelial function and oxidative stress than mean glucose in normal and type 2 diabetic patients. Diabetes 2008, 57, 1349–1354. [Google Scholar] [CrossRef]
  244. Vijayalingam, S.; Parthiban, A.; Shanmugasundaram, K.R.; Mohan, V. Abnormal antioxidant status in impaired glucose tolerance and non-insulin-dependent diabetes mellitus. Diabet. Med. 1996, 13, 715–719. [Google Scholar] [CrossRef]
  245. Bast, A.; Wolf, G.; Oberbaumer, I.; Walther, R. Oxidative and nitrosative stress induces peroxiredoxins in pancreatic beta cells. Diabetologia 2002, 45, 867–876. [Google Scholar] [CrossRef] [Green Version]
  246. Esposito, K.; Nappo, F.; Marfella, R.; Giugliano, G.; Giugliano, F.; Ciotola, M.; Quagliaro, L.; Ceriello, A.; Giugliano, D. Inflammatory cytokine concentrations are acutely increased by hyperglycemia in humans: Role of oxidative stress. Circulation 2002, 106, 2067–2072. [Google Scholar] [CrossRef] [PubMed]
  247. Heilbronn, L.K.; Campbell, L.V. Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr. Pharm. Des. 2008, 14, 1225–1230. [Google Scholar] [CrossRef] [PubMed]
  248. Matulewicz, N.; Karczewska-Kupczewska, M. Insulin resistance and chronic inflammation. Postepy. Hig. Med. Dosw. 2016, 70, 1245–1258. [Google Scholar]
  249. Bonamichi, B.; Lee, J. Unusual Suspects in the Development of Obesity-Induced Inflammation and Insulin Resistance: NK cells, iNKT cells, and ILCs. Diabetes Metab. J. 2017, 41, 229–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Chen, Z.; Yu, R.; Xiong, Y.; Du, F.; Zhu, S. A vicious circle between insulin resistance and inflammation in nonalcoholic fatty liver disease. Lipids. Health Dis. 2017, 16. [Google Scholar] [CrossRef] [PubMed]
  251. Ricci, E.; Cipriani, S.; Chiaffarino, F.; Malvezzi, M.; Parazzini, F. Effects of soy isoflavones and genistein on glucose metabolism in perimenopausal and postmenopausal non-Asian women: A meta-analysis of randomized controlled trials. Menopause 2010, 17, 1080–1086. [Google Scholar] [CrossRef] [PubMed]
  252. Shrime, M.G.; Bauer, S.R.; McDonald, A.C.; Chowdhury, N.H.; Coltart, C.E.; Ding, E.L. Flavonoid-rich cocoa consumption affects multiple cardiovascular risk factors in a meta-analysis of short-term studies. J. Nutr. 2011, 141, 1982–1988. [Google Scholar] [CrossRef] [PubMed]
  253. Curtis, P.J.; Sampson, M.; Potter, J.; Dhatariya, K.; Kroon, P.A.; Cassidy, A. Chronic ingestion of flavan-3-ols and isoflavones improves insulin sensitivity and lipoprotein status and attenuates estimated 10-year CVD risk in medicated postmenopausal women with type 2 diabetes: A 1-year, double-blind, randomized, controlled trial. Diabetes Care 2012, 35, 226–232. [Google Scholar] [CrossRef]
  254. Grassi, D.; Necozione, S.; Lippi, C.; Croce, G.; Valeri, L.; Pasqualetti, P.; Desideri, G.; Blumberg, J.B.; Ferri, C. Cocoa reduces blood pressure and insulin resistance and improves endothelium-dependent vasodilation in hypertensives. Hypertension 2005, 46, 398–405. [Google Scholar] [CrossRef]
  255. Grassi, D.; Lippi, C.; Necozione, S.; Desideri, G.; Ferri, C. Short-term administration of dark chocolate is followed by a significant increase in insulin sensitivity and a decrease in blood pressure in healthy persons. Am. J. Clin. Nutr. 2005, 81, 611–614. [Google Scholar] [CrossRef]
  256. Desideri, G.; Kwik-Uribe, C.; Grassi, D.; Necozione, S.; Ghiadoni, L.; Mastroiacovo, D.; Raffaele, A.; Ferri, L.; Bocale, R.; Lechiara, M.C.; et al. Benefits in cognitive function, blood pressure, and insulin resistance through cocoa flavanol consumption in elderly subjects with mild cognitive impairment: The Cocoa, Cognition, and Aging (CoCoA) study. Hypertension 2012, 60, 794–801. [Google Scholar] [CrossRef] [PubMed]
  257. Weseler, A.R.; Geraets, L.; Moonen, H.J.; Manders, R.J.; van Loon, L.J.; Pennings, H.J.; Wouters, E.F.; Bast, A.; Hageman, G.J. Poly (ADP-ribose) polymerase-1-inhibiting flavonoids attenuate cytokine release in blood from male patients with chronic obstructive pulmonary disease or type 2 diabetes. J. Nutr. 2009, 139, 952–957. [Google Scholar] [CrossRef] [PubMed]
  258. Hoensch, H.P.; Oertel, R. The value of flavonoids for the human nutrition: Short review and perspectives. Clin. Nutr. Exp. 2015, 3, 8–14. [Google Scholar] [CrossRef] [Green Version]
  259. Perez-Cano, F.J.; Castell, M. Flavonoids, Inflammation and Immune System. Nutrients 2016, 8, 659. [Google Scholar] [CrossRef] [PubMed]
  260. Galleano, M.; Calabro, V.; Prince, P.D.; Litterio, M.C.; Piotrkowski, B.; Vazquez-Prieto, M.A.; Miatello, R.M.; Oteiza, P.I.; Fraga, C.G. Flavonoids and metabolic syndrome. Ann. N. Y. Acad. Sci. 2012, 1259, 87–94. [Google Scholar] [CrossRef] [PubMed]
  261. Blumberg, J.B.; Ding, E.L.; Dixon, R.; Pasinetti, G.M.; Villarreal, F. The science of cocoa flavanols: Bioavailability, emerging evidence, and proposed mechanisms. Adv. Nutr. 2014, 5, 547–549. [Google Scholar] [CrossRef] [PubMed]
  262. Bohn, S.K.; Croft, K.D.; Burrows, S.; Puddey, I.B.; Mulder, T.P.; Fuchs, D.; Woodman, R.J.; Hodgson, J.M. Effects of black tea on body composition and metabolic outcomes related to cardiovascular disease risk: A randomized controlled trial. Food Funct. 2014, 5, 1613–1620. [Google Scholar] [CrossRef] [PubMed]
  263. Mellor, D.D.; Sathyapalan, T.; Kilpatrick, E.S.; Beckett, S.; Atkin, S.L. High-cocoa polyphenol-rich chocolate improves HDL cholesterol in Type 2 diabetes patients. Diabet. Med. 2010, 27, 1318–1321. [Google Scholar] [CrossRef] [PubMed]
  264. Polagruto, J.A.; Wang-Polagruto, J.F.; Braun, M.M.; Lee, L.; Kwik-Uribe, C.; Keen, C.L. Cocoa flavanol-enriched snack bars containing phytosterols effectively lower total and low-density lipoprotein cholesterol levels. J. Am. Diet. Assoc. 2006, 106, 1804–1813. [Google Scholar] [CrossRef] [PubMed]
  265. Gardner, C.D.; Newell, K.A.; Cherin, R.; Haskell, W.L. The effect of soy protein with or without isoflavones relative to milk protein on plasma lipids in hypercholesterolemic postmenopausal women. Am. J. Clin. Nutr. 2001, 73, 728–735. [Google Scholar] [CrossRef] [Green Version]
  266. Wilson, T.; Luebke, J.L.; Morcomb, E.F.; Carrell, E.J.; Leveranz, M.C.; Kobs, L.; Schmidt, T.P.; Limburg, P.J.; Vorsa, N.; Singh, A.P. Glycemic responses to sweetened dried and raw cranberries in humans with type 2 diabetes. J. Food Sci. 2010, 75, 218–223. [Google Scholar] [CrossRef] [PubMed]
  267. Hokayem, M.; Blond, E.; Vidal, H.; Lambert, K.; Meugnier, E.; Feillet-Coudray, C.; Coudray, C.; Pesenti, S.; Luyton, C.; Lambert-Porcheron, S.; et al. Grape polyphenols prevent fructose-induced oxidative stress and insulin resistance in first-degree relatives of type 2 diabetic patients. Diabetes Care 2013, 36, 1454–1461. [Google Scholar] [CrossRef] [PubMed]
  268. de Bock, M.; Derraik, J.G.; Brennan, C.M.; Biggs, J.B.; Morgan, P.E.; Hodgkinson, S.C.; Hofman, P.L.; Cutfield, W.S. Olive (Olea europaea L.) leaf polyphenols improve insulin sensitivity in middle-aged overweight men: A randomized, placebo-controlled, crossover trial. PLoS ONE 2013, 8. [Google Scholar] [CrossRef] [PubMed]
  269. Ebrahimpour Koujan, S.; Gargari, B.P.; Mobasseri, M.; Valizadeh, H.; Asghari-Jafarabadi, M. Effects of Silybum marianum (L.) Gaertn. (silymarin) extract supplementation on antioxidant status and hs-CRP in patients with type 2 diabetes mellitus: A randomized, triple-blind, placebo-controlled clinical trial. Phytomedicine 2015, 22, 290–296. [Google Scholar] [CrossRef] [PubMed]
  270. Sahebkar, A. Effects of quercetin supplementation on lipid profile: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2017, 57, 666–676. [Google Scholar] [CrossRef] [PubMed]
  271. Pfeuffer, M.; Auinger, A.; Bley, U.; Kraus-Stojanowic, I.; Laue, C.; Winkler, P.; Rufer, C.E.; Frank, J.; Bosch-Saadatmandi, C.; Rimbach, G.; et al. Effect of quercetin on traits of the metabolic syndrome, endothelial function and inflammation in men with different APOE isoforms. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 403–409. [Google Scholar] [CrossRef] [PubMed]
  272. Dower, J.I.; Geleijnse, J.M.; Gijsbers, L.; Schalkwijk, C.; Kromhout, D.; Hollman, P.C. Supplementation of the Pure Flavonoids Epicatechin and Quercetin Affects Some Biomarkers of Endothelial Dysfunction and Inflammation in (Pre)Hypertensive Adults: A Randomized Double-Blind, Placebo-Controlled, Crossover Trial. J. Nutr. 2015, 145, 1459–1463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Boots, A.W.; Drent, M.; de Boer, V.C.; Bast, A.; Haenen, G.R. Quercetin reduces markers of oxidative stress and inflammation in sarcoidosis. Clin. Nutr. 2011, 30, 506–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Brull, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Muller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Stehle, P.; et al. No effects of quercetin from onion skin extract on serum leptin and adiponectin concentrations in overweight-to-obese patients with (pre-)hypertension: A randomized double-blinded, placebo-controlled crossover trial. Eur. J. Nutr. 2017, 56, 2265–2275. [Google Scholar] [CrossRef]
  275. Guo, Y.; Mah, E.; Davis, C.G.; Jalili, T.; Ferruzzi, M.G.; Chun, O.K.; Bruno, R.S. Dietary fat increases quercetin bioavailability in overweight adults. Mol. Nutr. Food Res. 2013, 57, 896–905. [Google Scholar] [CrossRef]
  276. Heinz, S.A.; Henson, D.A.; Nieman, D.C.; Austin, M.D.; Jin, F. A 12-week supplementation with quercetin does not affect natural killer cell activity, granulocyte oxidative burst activity or granulocyte phagocytosis in female human subjects. Br. J. Nutr. 2010, 104, 849–857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Egert, S.; Boesch-Saadatmandi, C.; Wolffram, S.; Rimbach, G.; Muller, M.J. Serum lipid and blood pressure responses to quercetin vary in overweight patients by apolipoprotein E genotype. J. Nutr. 2010, 140, 278–284. [Google Scholar] [CrossRef] [PubMed]
  278. Javadi, F.; Ahmadzadeh, A.; Eghtesadi, S.; Aryaeian, N.; Zabihiyeganeh, M.; Rahimi Foroushani, A.; Jazayeri, S. The Effect of Quercetin on Inflammatory Factors and Clinical Symptoms in Women with Rheumatoid Arthritis: A Double-Blind, Randomized Controlled Trial. J. Am. Coll. Nutr. 2017, 36, 9–15. [Google Scholar] [CrossRef] [PubMed]
  279. Talirevic, E.; Jelena, S. Quercetin in the treatment of dyslipidemia. Med. Arch. 2012, 66, 87–88. [Google Scholar] [CrossRef]
  280. Gutierrez-Salmean, G.; Meaney, E.; Lanaspa, M.A.; Cicerchi, C.; Johnson, R.J.; Dugar, S.; Taub, P.; Ramirez-Sanchez, I.; Villarreal, F.; Schreiner, G.; et al. A randomized, placebo-controlled, double-blind study on the effects of (-)-epicatechin on the triglyceride/HDLc ratio and cardiometabolic profile of subjects with hypertriglyceridemia: Unique in vitro effects. Int. J. Cardiol. 2016, 223, 500–506. [Google Scholar] [CrossRef] [PubMed]
  281. Homayouni, F.; Haidari, F.; Hedayati, M.; Zakerkish, M.; Ahmadi, K. Hesperidin Supplementation Alleviates Oxidative DNA Damage and Lipid Peroxidation in Type 2 Diabetes: A Randomized Double-Blind Placebo-Controlled Clinical Trial. Phytother. Res. 2017, 31, 1539–1545. [Google Scholar] [CrossRef] [PubMed]
  282. Jung, U.J.; Kim, H.J.; Lee, J.S.; Lee, M.K.; Kim, H.O.; Park, E.J.; Kim, H.K.; Jeong, T.S.; Choi, M.S. Naringin supplementation lowers plasma lipids and enhances erythrocyte antioxidant enzyme activities in hypercholesterolemic subjects. Clin. Nutr. 2003, 22, 561–568. [Google Scholar] [CrossRef]
  283. Squadrito, F.; Marini, H.; Bitto, A.; Altavilla, D.; Polito, F.; Adamo, E.B.; D’Anna, R.; Arcoraci, V.; Burnett, B.P.; Minutoli, L.; et al. Genistein in the metabolic syndrome: Results of a randomized clinical trial. J. Clin. Endocrinol. Metab. 2013, 98, 3366–3374. [Google Scholar] [CrossRef]
  284. Xiao, L.Z.; Huang, Z.; Ma, S.C.; Zen, Z.; Luo, B.; Lin, X.; Xu, X. [Study on the effect and mechanism of puerarin on the size of infarction in patients with acute myocardial infarction]. Zhongguo Zhong Xi Yi Jie He Za Zhi 2004, 24, 790–792. [Google Scholar]
  285. Shi, W.G.; Qu, L.; Wang, J.W. [Study on interventing effect of puerarin on insulin resistance in patients with coronary heart disease]. Zhongguo Zhong Xi Yi Jie He Za Zhi 2002, 22, 21–24. [Google Scholar]
  286. Villa, P.; Costantini, B.; Suriano, R.; Perri, C.; Macri, F.; Ricciardi, L.; Panunzi, S.; Lanzone, A. The differential effect of the phytoestrogen genistein on cardiovascular risk factors in postmenopausal women: Relationship with the metabolic status. J. Clin. Endocrinol. Metab. 2009, 94, 552–558. [Google Scholar] [CrossRef] [PubMed]
  287. Marini, H.; Bitto, A.; Altavilla, D.; Burnett, B.P.; Polito, F.; Di Stefano, V.; Minutoli, L.; Atteritano, M.; Levy, R.M.; Frisina, N.; et al. Efficacy of genistein aglycone on some cardiovascular risk factors and homocysteine levels: A follow-up study. Nutr. Metab. Cardiovasc. Dis. 2010, 20, 332–340. [Google Scholar] [CrossRef] [PubMed]
  288. de Ruijter, J.; Valstar, M.J.; Narajczyk, M.; Wegrzyn, G.; Kulik, W.; Ijlst, L.; Wagemans, T.; van der Wal, W.M.; Wijburg, F.A. Genistein in Sanfilippo disease: A randomized controlled crossover trial. Ann. Neurol. 2012, 71, 110–120. [Google Scholar] [CrossRef] [PubMed]
  289. Egert, S.; Bosy-Westphal, A.; Seiberl, J.; Kurbitz, C.; Settler, U.; Plachta-Danielzik, S.; Wagner, A.E.; Frank, J.; Schrezenmeir, J.; Rimbach, G.; et al. Quercetin reduces systolic blood pressure and plasma oxidised low-density lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: A double-blinded, placebo-controlled cross-over study. Br. J. Nutr. 2009, 102, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  290. Shi, Y.; Williamson, G. Quercetin lowers plasma uric acid in pre-hyperuricaemic males: A randomised, double-blinded, placebo-controlled, cross-over trial. Br. J. Nutr. 2016, 115, 800–806. [Google Scholar] [CrossRef] [PubMed]
  291. Mielgo-Ayuso, J.; Barrenechea, L.; Alcorta, P.; Larrarte, E.; Margareto, J.; Labayen, I. Effects of dietary supplementation with epigallocatechin-3-gallate on weight loss, energy homeostasis, cardiometabolic risk factors and liver function in obese women: Randomised, double-blind, placebo-controlled clinical trial. Br. J. Nutr. 2014, 111, 1263–1271. [Google Scholar] [CrossRef] [PubMed]
  292. Brown, A.L.; Lane, J.; Coverly, J.; Stocks, J.; Jackson, S.; Stephen, A.; Bluck, L.; Coward, A.; Hendrickx, H. Effects of dietary supplementation with the green tea polyphenol epigallocatechin-3-gallate on insulin resistance and associated metabolic risk factors: Randomized controlled trial. Br. J. Nutr. 2009, 101, 886–894. [Google Scholar] [CrossRef]
  293. Boschmann, M.; Thielecke, F. The effects of epigallocatechin-3-gallate on thermogenesis and fat oxidation in obese men: A pilot study. J. Am. Coll. Nutr. 2007, 26, 389–395. [Google Scholar] [CrossRef]
  294. Meshitsuka, S.; Shingaki, S.; Hotta, M.; Goto, M.; Kobayashi, M.; Ukawa, Y.; Sagesaka, Y.M.; Wada, Y.; Nojima, M.; Suzuki, K. Phase 2 trial of daily, oral epigallocatechin gallate in patients with light-chain amyloidosis. Int. J. Hematol. 2017, 105, 295–308. [Google Scholar] [CrossRef]
  295. Most, J.; Timmers, S.; Warnke, I.; Jocken, J.W.; van Boekschoten, M.; de Groot, P.; Bendik, I.; Schrauwen, P.; Goossens, G.H.; Blaak, E.E. Combined epigallocatechin-3-gallate and resveratrol supplementation for 12 wk increases mitochondrial capacity and fat oxidation, but not insulin sensitivity, in obese humans: A randomized controlled trial. Am. J. Clin. Nutr. 2016, 104, 215–227. [Google Scholar] [CrossRef]
  296. Haidari, F.; Heybar, H.; Jalali, M.T.; Ahmadi Engali, K.; Helli, B.; Shirbeigi, E. Hesperidin supplementation modulates inflammatory responses following myocardial infarction. J. Am. Coll. Nutr. 2015, 34, 205–211. [Google Scholar] [CrossRef] [PubMed]
  297. Demonty, I.; Lin, Y.; Zebregs, Y.E.; Vermeer, M.A.; van der Knaap, H.C.; Jakel, M.; Trautwein, E.A. The citrus flavonoids hesperidin and naringin do not affect serum cholesterol in moderately hypercholesterolemic men and women. J. Nutr. 2010, 140, 1615–1620. [Google Scholar] [CrossRef] [PubMed]
  298. Rizza, S.; Muniyappa, R.; Iantorno, M.; Kim, J.A.; Chen, H.; Pullikotil, P.; Senese, N.; Tesauro, M.; Lauro, D.; Cardillo, C.; et al. Citrus polyphenol hesperidin stimulates production of nitric oxide in endothelial cells while improving endothelial function and reducing inflammatory markers in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 2011, 96, 782–792. [Google Scholar] [CrossRef] [PubMed]
  299. Delgadillo, V.; O’Callaghan Mdel, M.; Artuch, R.; Montero, R.; Pineda, M. Genistein supplementation in patients affected by Sanfilippo disease. J. Inherit. Metab. Dis. 2011, 34, 1039–1044. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Inflammation and metabolic disorders.
Figure 1. Inflammation and metabolic disorders.
Ijms 20 04957 g001
Figure 2. Chemical structure of most relevant flavonoids with therapeutic effects in metabolic syndrome by targeting inflammatory pathways.
Figure 2. Chemical structure of most relevant flavonoids with therapeutic effects in metabolic syndrome by targeting inflammatory pathways.
Ijms 20 04957 g002
Figure 3. Underlying anti-inflammatory pathways involved in therapeutic effects of flavonoids in metabolic syndrome. NF-κB, nuclear factor-κB; IκB, inhibitor of κB; AMPK, activated protein kinase; SIRT1, Sirtuin 1; JNK, c-Jun N-terminal kinase; KLF2, Kruppel-like factor 2; AP-1, activator protein 1; Nrf2, nuclear factor erythroid 2-related factor 2; PLA2, phospholipase A2; COX, cyclooxygenase; eNOS, endothelial nitric oxide synthase; TNF-α; tumor necrosis factor-α; IL, interleukin, ROS, reactive oxygen species; red↓, decrease; all other ↓, direction of events.
Figure 3. Underlying anti-inflammatory pathways involved in therapeutic effects of flavonoids in metabolic syndrome. NF-κB, nuclear factor-κB; IκB, inhibitor of κB; AMPK, activated protein kinase; SIRT1, Sirtuin 1; JNK, c-Jun N-terminal kinase; KLF2, Kruppel-like factor 2; AP-1, activator protein 1; Nrf2, nuclear factor erythroid 2-related factor 2; PLA2, phospholipase A2; COX, cyclooxygenase; eNOS, endothelial nitric oxide synthase; TNF-α; tumor necrosis factor-α; IL, interleukin, ROS, reactive oxygen species; red↓, decrease; all other ↓, direction of events.
Ijms 20 04957 g003
Table 1. In vitro studies on polyphenols, improving metabolic disorders via ameliorating inflammatory pathways.
Table 1. In vitro studies on polyphenols, improving metabolic disorders via ameliorating inflammatory pathways.
PolyphenolsCell LineMechanism of ActionReferences
Flavonols (kaempferol, quercetin, and galangin)Human aortic endothelial cellsInhibiting the expression of intercellular adhesion molecule 1 (ICAM-1) and E-selectin[194]
Hydroxyl flavones (apigenin and chrysin)Human aortic endothelial cellsInhibiting the expression of ICAM-1 and E-selectin[194]
QuercetinHuman endothelial cellsReducing the transcription factor activator protein (AP)-1 and the expression of ICAM-1[195]
Human umbilical vein endothelial cells (HUVECs)Reducing the adhesion molecule and monocyte chemotactic protein-1 (MCP-1) gene expression[201]
Aortic endothelial cellsInhibiting the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) & AP-1 DNA-binding activity[202]
ProanthocyanidinsHuman endothelial cellsSuppressing the vascular cell adhesion molecule-1 (VCAM-1) expression[196]
GenisteinHuman endothelial cellsInhibiting the monocytes adhesion[197,198]
Cocoa extract (CE), epicatechin (EC), and isoquercitrin (IQ)Macrophages1-CE, EC, and IQ reduced tumor necrosis factor- alpha (TNF-α) levels
2-CE and EC reduced the messenger RNA (mRNA) expression of TNF-α, interleukin (IL)-1α, and IL-6
[200]
AnthocyaninsHepG2 cellsDecreasing the C-reactive protein production[203]
Porcine iliac artery endothelial cellsSuppressing the VCAM-1 expression[203]
Juice from bergamot (Citrus bergamia L.) rich in flavonoids (neohesperidin, naringin, melitidin, meoeriocitrin, and hesperetin)THP-1 cellsInhibiting the NF-kB activation
Sirtuin 1 (SIRT1) activation
[204]
Baicalin, baicalein, or wogoninThe nucleus of HUVECsInhibiting vascular inflammation through decreasing the p65 NF-κB expression[205]
NaringeninHUVECsAnti-inflammatory
Anti-atherosclerotic
Inhibiting monocyte-endothelium adhesion
Decreasing chemokines levels and NF-κB nuclear translocation via reduced phosphorylation of IKKα/β, IκB-α, and NF-κB
[206]
Puerarin, nobiletin, quercetin, quercetin 7-O-sialic acid, luteolin, and apigeninHUVECsInhibiting the NK-κB pathway[207,208,209,210,211]
Hesperetin-3′-sulgate, hesperetin-3′-glucur-onide, and naringenin-4′-glucuronideHUVECsReducing the adhesion of monocytes
Inhibiting several chemokines due to NF-κB inhibition
[212]
Phytoestrogen extracts from Glycine max (soybean), genistein, formononetin, biochanin A, and daidzeinHUVECsReducing the adhesion of monocytes
Inhibiting several chemokines due to NF-κB inhibition
[213]
Vitexicarpin, kaempferol, pelargonidin, epigallocatechin-3-gallate, and biochanin AHUVECsInhibiting the NF-κB[214,215,216,217,218]
A black rice (Oryza sativa L.) polar extract, highly rich in anthocyaninRaw 264.7 macrophage cellsInhibiting NF-κB and AP-1 translocation into the nucleus[219]
Dimocarpus longan Lour. flowersRaw 264.7 macrophage cellsSuppressing the NF-κB and AP-1 signaling pathways[220]
Cerbera manghas L. leaves, rich in flavonoidsRaw 264.7 macrophage cellsSuppressing the c-Jun N-terminal kinase in AP-1 pathway[221]
KaempferolRaw 264.7 macrophage cellsInhibiting the AP-1 activity[221]
NobiletinHepG2Inhibiting the AP-1 activity[222]
Berry anthocyaninsRaw 264.7 macrophage cellsDecreasing reactive oxygen species (ROS) levels through an nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent mechanism[223]
Prunella vulgaris L. var. lilacina, rich in flavonoidsRaw 264.7 macrophage cellsIncreasing the Heme oxygenase (HO)-1 protein expression through a mechanism involving phosphoinositide 3-kinases (PI3K)/Nrf2 pathways[224]
IsovitexinRaw 264.7 macrophage cellsIncreasing the HO-1 expression levels
Increasing the activation of Nrf2
[225]
Luteolin and luteolin-7-O-glucosideRaw 264.7 macrophage cellsIncreasing Nrf2 activation through the regulation of p38 and c-Jun N-terminal kinase (JNK) signaling pathway[226]
Grape seed proanthocyanidin extractsHUVECsInducing the endothelial nitric oxide synthase (eNOS) expression through the increase of Kruppel-like factor 2 (KLF2) expression
Increasing the 5’ AMP-activated protein kinase (AMPK) phosphorylation and SIRT1 protein level
[227]
Table 2. Clinical trials of pure flavonoids used in the treatment of metabolic disorder-associated diseases.
Table 2. Clinical trials of pure flavonoids used in the treatment of metabolic disorder-associated diseases.
FlavonoidDoseTrial Type and PatientsNo. of PatientsDuration of TreatmentSignificant ResultsReference
Quercetin150 mg/dayPlacebo-controlled, randomized, double-blind, cross-over study in healthy volunteers with apoE genotype 3/3, 3/4 and 4/4498 weeks↓Waist circumference, postprandial systolic blood pressure (BP), and postprandial triglyceride (TG) concentrations, ↑high-density lipoprotein (HDL)-cholesterol concentrations and levels of tumor necrosis factor-α (TNF-α)[271]
160 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in healthy (pre)hypertensive men and women, aged 40–80 years, with a systolic BP = 125–160 mm Hg264 weeksNo effect on flow-mediated dilation, insulin resistance, or other cardio vascular diseases (CVD) risk factors[272]
2000 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in non-smoking, untreated sarcoidosis patients, matched for age and gender181 day↑Total plasma antioxidant capacity; ↓markers of oxidative stress and inflammation in the blood of sarcoidosis patients[273]
160 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in healthy (pre)hypertensive volunteers (40–80 years)374 weeks↓Levels of sE-selectin, interleukin (IL)-1β, ↓endothelial dysfunction[237]
150 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight/obese subjects aged 25–65 years with metabolic syndrome traits936 weeks↓Serum HDL-cholesterol concentrations, ↓oxidized LDL[289]
162 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight-to-obese patients with pre- and stage 1 hypertension686 weeksNo significant effects on parameters tested[274]
1095 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight/obese men and postmenopausal women91 dayDietary fat increased the levels of methylated quercetin metabolites[275]
150 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight-obese patients aged 25–65 years with metabolic syndrome traits in relation to (apo) E genotype935 weeks↓Systolic BP in the apoE3 group, ↓serum HDL cholesterol and apoA1, ↑LDL:HDL cholesterol ratio, ↓plasma oxidized LDL and tumor necrosis factor-alpha in the apoE3 and apoE4 groups[277]
500 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in healthy volunteers (19–60 years) with higher plasma uric acid concentration224 weeks↓Plasma uric acid concentrations[290]
-Placebo-controlled, randomized, double-blind, clinical study in healthy patients with dyslipidemia2008 weeks↓Average cholesterol, triglycerides, cholesterol, triglyceride and low-density lipoprotein (LDL) values with parallel increase in HDL[279]
500 mg/day;1000 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in healthy female subjects (aged 30–79 years)12012 weeks↑Plasma quercetin at both doses[276]
500 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in women with rheumatoid arthritis (RA)508 Weeks↓Early morning stiffness (EMS, tender joint count (TJC), morning pain, and after-activity pain, ↓28-joint disease activity score (DAS-28) and health assessment questionnaire (HAQ) scores, ↓plasma hs-TNF-α level[278]
Epicatechin100 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in healthy (pre)hypertensive volunteers (40–80 years)374 weeks↓Endothelial dysfunction[237]
100 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in subjects with hypertriglyceridemia304 weeksFavorable effects on glycemia homeostasis, lipid profile and systemic inflammation[280]
Epigallocatechin-3-gallate (EGCG)300 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in obese, pre-menopausal Caucasian women8312 weeksNo significant effects on parameters tested[291]
400 m/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight/obese male subjects, aged 40–65 years468 weeks↓Diastolic BP[292]
300 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight men63 days↓Respiratory quotient (RQ)[293]
1890 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with light-chain (AL) amyloidosis5724 weeks↓Urinary albumin level[294]
282 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in overweight and obese subjects3812 weeks↓Visceral adipose tissue mass, ↑increased oxidative capacity in permeabilized muscle fibers, ↓fasting and postprandial respiratory quotient, ↓increase in plasma triacylglycerol concentrations[295]
Hesperidin600 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with myocardial infarction754 weeks↓Serum E-selectin levels, ↑adiponectin and HDL-C concentrations, ↑interleukin (IL)-6, high sensitivity C-reactive protein (hs-CRP), leptin, and lipid profile[296]
800 mg/dayPlacebo-controlled, randomized, double-blind, trial in moderately hypercholesterolemic men and women1364 weeksNo significant effects on parameters tested[297]
500 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with type 2 diabetes646 weeks↑Total antioxidant capacity (TAC), ↓serum fructosamine, ↓ malondialdehyde (MDA), ↑8-hydroxy-2’ -deoxyguanosine (8-OHDG)[281]
500 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with metabolic syndrome243 weeks↑Flow-mediated, ↓high-sensitivity C-reactive protein, serum amyloid A protein, soluble E-selectin[298]
Naringin500 mg/dayPlacebo-controlled, randomized, double-blind, trial in moderately hypercholesterolemic men and women1364 weeksNo significant effects on parameters tested[297]
400 mg/dayTwo group of patients: hypercholesterolemic and health608 weeks↓The plasma total cholesterol, ↓low-density lipoprotein cholesterol, ↓Apo B levels, ↑erythrocyte superoxide dismutase and catalase[282]
Genistein54 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in postmenopausal women with metabolic syndrome (MetS)1201 year↓Fasting glucose, ↓fasting insulin, ↓homeostasis model assessment of insulin resistance (HOMA-IR) ↑HDL-C, ↑adiponectin, ↓total cholesterol, ↓LDL-C, ↓triglycerides, ↓visfatin, ↓homocysteine, ↓systolic and diastolic BP[283]
10 mg/kg/dayPlacebo-controlled, randomized, double-blind,, clinical study in patients with mucopolysaccharidosis type III (MPS III) (Sanfilippo disease)301 year↓Urinary excretion of total glycosaminoglycans (GAGs), ↓plasma hydrogen sulfide (HS) concentrations[288]
5 mg/kg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with confirmed diagnosis of MPS III (age range 2.8–19 years)191 year↓Recurrence of infections and gastrointestinal symptoms, ↑ skin texture and hair morphology[299]
54 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in normoinsulinemic and hyperinsulinemic patients.5024 weeks↓Insulin basal values,↑homeostasis model index of insulin sensitivity, ↑fasting glucose levels, ↓fasting insulin, fasting C-peptide, ↑ fractional hepatic insulin extraction was shown, ↑ HDL-C levels, ↑endothelium-dependent and -independent dilatation[286]
54 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in postmenopausal women with low bone mass1383 years↓Fasting glucose, ↓insulin, ↓HOMA-IR, ↓fibrinogen, ↓homocysteine[287]
Puerarin500 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with acute myocardial infarction612 weeks↓Free fatty acids (FFAs), ↓matrix metallopeptidase 9 (MMP-9), ↓c-reactive protein (CRP), ↓ infarction size[284]
500 mg/dayPlacebo-controlled, randomized, double-blind, clinical study in patients with coronary heart disease763 weeks↑Plasminogen activator inhibitor-1 (PAI-1), ↓insulin sensitivity index (ISI), ↓high density lipoprotein cholesterol (HDL-C), ↓tissue plasminogen activator (tPA), ↓FINS level, ↑ ISI ↑Total cholesterol (TC), ↑triglycerides (TG), ↑low-density lipoprotein (LDL), ↑fasting plasma glucose (FPG), ↑fasting insulin (FINS)[285]
↓: Decrease; ↑: Increase.

Share and Cite

MDPI and ACS Style

Farzaei, M.H.; Singh, A.K.; Kumar, R.; Croley, C.R.; Pandey, A.K.; Coy-Barrera, E.; Kumar Patra, J.; Das, G.; Kerry, R.G.; Annunziata, G.; et al. Targeting Inflammation by Flavonoids: Novel Therapeutic Strategy for Metabolic Disorders. Int. J. Mol. Sci. 2019, 20, 4957. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20194957

AMA Style

Farzaei MH, Singh AK, Kumar R, Croley CR, Pandey AK, Coy-Barrera E, Kumar Patra J, Das G, Kerry RG, Annunziata G, et al. Targeting Inflammation by Flavonoids: Novel Therapeutic Strategy for Metabolic Disorders. International Journal of Molecular Sciences. 2019; 20(19):4957. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20194957

Chicago/Turabian Style

Farzaei, Mohammad Hosein, Amit Kumar Singh, Ramesh Kumar, Courtney R. Croley, Abhay K. Pandey, Ericsson Coy-Barrera, Jayanta Kumar Patra, Gitishree Das, Rout George Kerry, Giuseppe Annunziata, and et al. 2019. "Targeting Inflammation by Flavonoids: Novel Therapeutic Strategy for Metabolic Disorders" International Journal of Molecular Sciences 20, no. 19: 4957. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20194957

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