Next Article in Journal
Dipeptide Nitrile CD34 with Curcumin: A New Improved Combination Strategy to Synergistically Inhibit Rhodesain of Trypanosoma brucei rhodesiense
Previous Article in Journal
Alterations in Antibiotic Susceptibility of Staphylococcus aureus and Klebsiella pneumoniae in Dual Species Biofilms
Previous Article in Special Issue
sFgl2-Treg Positive Feedback Pathway Protects against Atherosclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease

Cardiff School of Biosciences, Cardiff University, Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(10), 8476; https://doi.org/10.3390/ijms24108476
Submission received: 9 April 2023 / Revised: 26 April 2023 / Accepted: 3 May 2023 / Published: 9 May 2023

Abstract

:
Atherosclerotic cardiovascular disease (ACVD) is the leading cause of death worldwide. Although current therapies, such as statins, have led to a marked reduction in morbidity and mortality from ACVD, they are associated with considerable residual risk for the disease together with various adverse side effects. Natural compounds are generally well-tolerated; a major recent goal has been to harness their full potential in the prevention and treatment of ACVD, either alone or together with existing pharmacotherapies. Punicalagin (PC) is the main polyphenol present in pomegranates and pomegranate juice and demonstrates many beneficial actions, including anti-inflammatory, antioxidant, and anti-atherogenic properties. The objective of this review is to inform on our current understanding of the pathogenesis of ACVD and the potential mechanisms underlying the beneficial actions of PC and its metabolites in the disease, including the attenuation of dyslipidemia, oxidative stress, endothelial cell dysfunction, foam cell formation, and inflammation mediated by cytokines and immune cells together with the regulation of proliferation and migration of vascular smooth muscle cells. Some of the anti-inflammatory and antioxidant properties of PC and its metabolites are due to their strong radical-scavenging activities. PC and its metabolites also inhibit the risk factors of atherosclerosis, including hyperlipidemia, diabetes mellitus, inflammation, hypertension, obesity, and non-alcoholic fatty liver disease. Despite the promising findings that have emerged from numerous in vitro, in vivo, and clinical studies, deeper mechanistic insights and large clinical trials are required to harness the full potential of PC and its metabolites in the prevention and treatment of ACVD.

1. Introduction

Cardiovascular disease (CVD) remains a leading cause of global morbidity and mortality, accounting for an estimated 17.9-million (31%) deaths according to the World Health Organization [1]. The burden from CVD continues to grow globally due to an increase in risk factors such as obesity, diabetes, high blood pressure, and the western lifestyle [1]. CVD was the cause of most deaths in the UK since the 1950s, but it was overtaken by cancer in 2011 [2]. In 2014, around 27.1% of deaths in the UK were due to CVD [3]. Atherosclerosis is a chronic immune and inflammatory disorder of medium and large arteries involving multiple cell types, including endothelial cells (EC), monocytes, macrophages, smooth muscle cells (SMC), T-lymphocytes, and mast cells, and it is the underlying cause of CVD [4,5,6]. The initiation of atherosclerosis typically starts with the accumulation of low-density lipoproteins (LDL) in the intima of arteries together with other risk factors, which promotes the recruitment of immune cells, including monocytes and lymphocytes. This subsequently leads to the formation of lipid-laden foam cells derived from macrophages and vascular smooth muscle cells (VSMCs) [1]. The retention of an excess amount of intracellular cholesterol in foam cells results in cellular dysfunction and subsequent stress responses, resulting in foam cell death and necrotic core formation [1]. Migrated VSMCs from the media to the intima secrete extracellular matrix (ECM) proteins that form the fibrous cap that covers the necrotic core, and thereby helps in plaque stabilization [1]. Atherosclerosis is a slow and complex disorder that develops over decades and progresses faster with age. Usually, atherosclerosis is asymptomatic until one of the arteries is blocked by thrombosis due to plaque rupture. Therefore, an early diagnosis of atherosclerosis can help via preventive care, including lifestyle modifications, exercise, and changes in diet and medical treatments, to reduce the clinical manifestations of atherosclerotic cardiovascular disease (ACVD), including myocardial infarction (MI), stroke, and ischemic heart diseases [7]. Even though pharmacotherapies, such as statins, have greatly lowered morbidity and mortality rates from ACVD, the persistent risk of primary and secondary major cardiovascular events that occurs after medication, as well as problems such as adverse side effects, has prompted research into alternate prevention and/or treatment approaches [1]. Much of the previous and ongoing research into potential preventative/therapeutic pathways for atherosclerosis have been motivated by the high prevalence of ACVD and the tremendous expense it imposes on healthcare systems [8,9].

2. Atherosclerosis

Previously, atherosclerosis was considered as a lipid storage disease. However, extensive research in the last three decades has revealed the significance of low-grade chronic inflammation, thereby explaining the molecular and cellular processes that contribute to atherogenesis [5,6]. Atherosclerosis starts when the various risk factors trigger EC dysfunction/activation that then causes these cells to secrete monocyte chemotactic protein-1 (MCP-1) and other chemokines, which attracts monocytes and other immune cells to the activated endothelium, followed by their adhesion to the endothelial cell surface [10]. Monocytes migrate to the site of inflammation via interactions between receptors on their cell surface and the adhesion molecules expressed on ECs, such as intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), E-selectin and, P-selectin [11], ultimately leading to the accumulation of monocytes in the subendothelial space and their subsequent differentiation into macrophages [8,11,12]. These then uptake oxidized LDL (oxLDL) and other modified LDL, which form in the subendothelial space, via endocytosis mediated by scavenger receptors on their cell surface, including lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), cluster of differentiation 36 (CD36), and scavenger receptor type 1 (SR-A1), along with other processes such as macropinocytosis and phagocytosis [12]. The modified LDL particles eventually move to the lysosomes as part of the endocytosis process to undergo enzymatic digestion [12]. The released free cholesterol is subsequently esterified to cholesteryl esters (CE) by acyl-CoA acyl transferase-1(ACAT1) and then transferred to the endoplasmic reticulum for storage as lipid droplets [12]. Macrophages with excess CE are called foam cells because of their “foamy” appearance [1]. The accumulation of cholesterol is toxic to the cells and initiates stress responses that ultimately causes them to undergo apoptosis and necrosis leading to the formation of a lipid-rich necrotic core [13]. The lipids in the necrotic core, such as cholesterol crystals, activate the inflammasome pathway leading to the secretion of interleukin (IL)-1β and IL-18, which, together with the production of other inflammatory mediators from various cells present in atherosclerotic plaque, cause a state of low-grade chronic inflammation [8,14,15]. In addition to the innate immune response, cells of the adaptive immune response (e.g., different subtypes of T-cells, B-cells, etc.) play key roles in regulating the chronic inflammation in atherosclerosis [4,5,6,16,17,18]. To counteract the detrimental changes and to maintain homeostasis, necrotic cells are cleared in the early stages of the disease by phagocytosis in a process called efferocytosis [19,20]. However, in advanced lesions, efferocytosis becomes ineffective as the plaques progress. Hence, there is an increase in the accumulation of apoptotic cells [1,19,20]. SMCs undergo a phenotypic shift (quiescent state to proliferative state) and migrate from the tunica media to the intima [1]. Some of migrated SMCs also become foam cells by the uptake of modified LDL. They also secrete ECM proteins, which contribute to the formation of a fibrous cap over the necrotic core [1]. As the formation of the fibrous cap progresses, a stable atherosclerotic lesion is formed [1]. Subsequently, ECM proteins are degraded by protease enzymes, particularly matrix metalloproteinases produced by macrophages, foam cells, and other cells during chronic inflammation [1]. This initiates the destabilization of the plaque, which ultimately leads to its rupture, thrombosis, and ACVD, including MI and cerebrovascular accidents [1,21]. Some of the risk factors for, and key cellular processes in, atherosclerosis are addressed below in more detail.

2.1. Risk Factors for Atherosclerosis

There are many risk factors for ACVD that are generally classified as modifiable and non-modifiable [22]. The former includes dyslipidemia, smoking, hypertension, diabetes, and obesity, whereas the latter includes age, male gender, and genetic predispositions such as familial hypercholesterolemia and Tangier disease [23]. Obesity, diabetes, and hypertension are all known to contribute to ACVD and increase its burden, but recent studies have also revealed an important role of non-alcoholic fatty liver disease (NAFLD) [24,25,26]. This is associated with an accumulation of lipids in the liver, which is caused by an imbalance between lipid uptake, storage, and utilization [27] in individuals who drink little or no alcohol [28]. Thus, NAFLD is characterized by an increase in the uptake of free fatty acids (FFA) by the liver and a decrease in their utilization for energy production, which initially leads to lipid accumulation that can eventually progress to nonalcoholic steatohepatitis (NASH) if untreated [25]. Atherosclerosis and NAFLD are closely linked through multiple mechanisms and risk factors such as diabetics, obesity, and metabolic syndromes [29]. Additionally, NAFLD is associated with increased oxidative stress and inflammation, which is also known to contribute to the development of atherosclerosis [30]. In addition, NAFLD is linked to changes in lipid metabolism that may have a direct role in the onset of atherosclerosis. Thus, triacylglycerol (TG) and LDL, known risk factors for atherosclerosis, are elevated in individuals with NAFLD, which is often also associated with reduced levels of the protective high-density lipoprotein (HDL) [31].

2.2. Role of Oxidative Stress in Atherosclerosis

An imbalance between the production of reactive nitrogen species (RNS) and reactive oxygen species (ROS), together with an absent or poor antioxidant system, leads to oxidative stress [32]. ROS can be of exogenous or endogenous origin, while the mitochondrial respiratory chain is the main endogenous source of ROS [33]. During the early stages of atherosclerotic lesion formation, enzymes such as nitric oxide synthase and xanthine oxidase contribute to oxidative stress [34], which is also associated with local inflammation, endothelial dysfunction, SMC proliferation, and plaque formation [35]. ROS also leads to the production of growth factors and mitogens by various cell types that then contributes to the stimulation of cell proliferation in early atherosclerotic lesions [36]. Oxidative stress also contributes to DNA instability, DNA mutations, dysfunction in the products of repair genes, and hyper-methylation [37]. Thus, ROS is considered as a major contributing factor in LDL oxidation and various signaling processes in the pathogenesis of atherosclerosis.

2.3. Role of Macrophages in Atherosclerosis

Macrophages are typically produced via differentiation of monocytes by macrophage-colony stimulating factors [38]. They are cells of the innate immune system that play pivotal roles in mounting an immunological reaction against foreign antigens or pathogens, including viruses and bacteria, and contribute to the regulation of inflammatory responses [39]. Several phenotypes of macrophages have been identified, with the two most common and well-documented being M1 (pro-inflammatory) and M2 (anti-inflammatory) [6,40]. M1 macrophages express several pro-inflammatory mediators, including IL-1, IL-6, ROS, RNS, and tumour necrosis factor (TNF)-α, while M2 macrophages are associated with the resolution of inflammation and express IL-10, the mannose receptor CD206, and arginase 1 [41]. Macrophages play a crucial role in atherosclerosis [8,42,43,44], where they are involved in all key processes, including foam cell formation, the development of lipid-rich necrotic core, the orchestration of the inflammatory response, and plaque rupture [12]. Excessive macrophage foam cell formation occurs during atherosclerosis because of an enhanced uptake of modified lipoproteins by scavenger receptors-mediated endocytosis, macropinocytosis and phagocytosis, and defective cholesterol efflux [1,12].

2.4. Role of Cytokines in Atherosclerosis

Cytokines are a class of small mediator proteins or glycoproteins that are secreted by many cells, including ECs, VSMCs, monocytes, macrophages, and T cells, in response to inflammation, infection, and other stimuli [6,8,45]. Cytokines are a complex series of proteins that comprise more than 100 released molecules that may be classified into various classes, including transforming growth factors, TNFs, colony-stimulating factors, ILs, interferons (IFNs) and chemokines [8,46]. The balance between pro-inflammatory and anti-inflammatory cytokines is crucial in the maintenance of cardiovascular health and in atherosclerosis, the balance is tipped towards the accumulation of pro-inflammatory cytokines [8,47]. As discussed above, cytokine-induced EC activation plays an important role in endothelial dysfunction and is followed by an increase in the recruitment and migration of immune cells into atherosclerotic sites [48]. The function of VSMCs is also regulated by cytokines that modulate their proliferation, migration, senescence, and phenotypic conversion [49].

2.5. Lysosomal Dysfunction in Atherosclerosis

As macrophages are involved in modified LDL uptake in early atherosclerotic lesions, their failure to digest the accumulated lipids contributes to the development and complexity of the disease [50]. Lysosomes play a crucial role in the maintenance of metabolic homeostasis of cells by degradation and sequestration of macromolecules [51], and lysosomal dysfunction is associated with sterile inflammation in atherosclerosis [52,53]. Macrophages develop features of lysosomal dysfunction following an exposure to atherogenic lipids [54]. Lysosomal stress can also activate transcription factor EB (TFEB), which acts as a main regulator of lysosomal biogenesis and function [55]. TFEB is regulated by multiple signaling pathways and can modulate several processes that are important in atherosclerosis, including lipophagy, autophagy, lipolysis, and inflammation [56,57,58,59,60,61]. Thus, lysosomal biogenesis in macrophages stimulated by TFEB may serve as a protective factor for atherosclerosis [62]. THP-1 macrophages treated with modified LDL develop lysosomal dysfunction [63], and mitochondrial ROS-induced lysosomal dysfunction promotes inflammation by contributing to M1 macrophage polarization [64]. Overall, lysosomal dysfunction is one of the multiple causes of atherosclerosis progression [54].

2.6. Reverse Cholesterol Transport and Atherosclerosis

Reverse cholesterol transport (RCT) is mediated by HDL, where this lipoprotein transports cholesterol from peripheral tissues back to the liver, where some of it is excreted via the bile system [12]. Hepatocytes and enterocytes are involved in the formation of nascent HDL that matures through the binding of phospholipids and free cholesterol effluxed from foam cells by ATP-binding cassette (ABC) transporters [12,65]. ABCA1 mediates cholesterol efflux to apolipoprotein (Apo) A1 present in HDL, whereas ABCG1 stimulates cellular cholesterol efflux to HDL [66]. Additionally, ABCA1 trafficking between the late endocytic vesicles and the cell surface is required for cholesterol efflux from endosomal/lysosomal compartments to lipid-free ApoA1 [67]. Furthermore, the intracellular sterol transporter ABCG1 stimulates cholesterol trafficking from the ER to the plasma membrane [68]. In addition to ABCA1 and ABCG1, the passive diffusion of cholesterol and scavenger receptor class B type 1 (SR-B1) enables the binding of lipids to HDL [69]. After the binding of cholesterol to HDL, the lecithin cholesteryl acyltransferase enzyme initiates esterification of the acquired cholesterol to form CE, forming mature HDL [70]. Hepatic lipase and endothelial lipase, respectively, mediate the remodeling of HDL particles via the hydrolysis of phospholipids and TG present in the lipoprotein [71]. The CE in the HDL core is transferred by cholesteryl ester transfer protein (CETP) to other lipoproteins, such as LDL, for removal by the liver through the low-density lipoprotein receptor (LDLr) and, ultimately, in the case of HDL, via SR-B1 [72]. CE is hydrolyzed in the liver, and free cholesterol is converted to bile acids that are used for the emulsification of lipids during intestinal digestion. The majority of bile is reabsorbed but some is lost from the body via the faeces [23,73].

3. Current Pharmacotherapies for Atherosclerosis

Statins, competitive inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, which catalyzes a rate-limiting step in the biosynthesis of cholesterol in the mevalonate pathway, are gold standard therapies against ACVD through reductions in plasma cholesterol levels [1,9,23]. The inhibition of the mevalonate pathway also decreases the levels of isoprenoid metabolites, geranylgeranyl pyrophosphate, and farnesyl pyrophosphate, that critically modify small-signaling G proteins that are involved in the regulation of numerous cellular functions, including survival, proliferation, and migration. Hence, these contribute to the pleiotropic actions of statins, such as anti-inflammatory actions and the attenuation of EC dysfunction [74,75]. However, statins are associated with various adverse side effects, such as myalgias, hepatic abnormalities, rhabdomyolysis, and diabetes in some cases [1,9,76]. Some emerging recent therapies target the intestinal absorption of dietary cholesterol (e.g., ezetimibe), plasma cholesterol levels (e.g., monoclonal antibodies against proprotein convertase subtilisin/kexin type-9, which prevent the degradation of LDLr, and bempedoic acid, which inhibits another enzyme in the cholesterol biosynthetic pathway), or inflammation (e.g., monoclonal antibody against the pro-inflammatory cytokine IL-1β and colchicine that inhibits the inflammasome pathway involved in the production of pro-inflammatory cytokines) [1,9]. Again, these agents have various issues, such as side effects (e.g., targeting inflammation makes individuals more prone to infections), costs associated with monoclonal antibodies as therapies, residual risk for disease, and non-compliance [1,9]. Many other promising agents have been unsuccessful in clinical targets because of side effects and off-target effects, including inhibitors of CETP, nicotinic acid, and its derivatives, such as niacin, and methotrexate [1,9,77]. Therefore, it is essential to find alternative preventive and therapeutic agents with better safety profile and less adverse side effects in the treatment ACVD.

4. Potential Nutraceutical Therapies for Atherosclerosis

One potential avenue being investigated for the prevention and treatment of ACVD is nutraceuticals or natural products with health benefits beyond their nutritional value, particularly those from phenolic-rich diets that are abundant in anti-oxidant and anti-inflammatory components [78,79,80,81]. The increased consumption of polyphenolic-rich fruits and vegetables has been linked to health benefits related to cardiovascular function. For example, it has been found that ACVD risk factors are reduced by 46% in individuals consuming polyphenol-rich diets [82]. Other studies have revealed that polyphenols can inhibit platelet aggregation [83], improve plasma lipid profile and inflammation markers [84], and help in maintaining endothelial function [85]. Pomegranate (Punica granatum) is rich in many polyphenol compounds, including anthocyanins and anthoxanthins, such as catechins, punicalagin (PC), ellagic acids (EA), gallic- and ellagi-tannins [86]. Both hydrolyzable ellagitannins (ET) and EA are potent antioxidants and are involved in the protection against atherogenesis [87]. PC is the main polyphenol ET in pomegranate and is responsible for its high antioxidant and anti-atherogenic activities [88]. Pomegranates cultivated in the Mediterranean, Middle East, India, China, Japan, and the United States have been reported as one of the fruits that contains the highest antioxidant, anti-atherogenic, anti-cancer, and anti-inflammatory components [89]. However, these potent effects are attributed to the highest concentration of polyphenols in pomegranates, including flavonoids, ET (e.g., PC, EA), and anthocyanins [90].

4.1. Bioavailability and Metabolism of Punicalagin and Its Metabolites

ET is a family of polyphenols present in nuts and fruits, such as walnuts, strawberries, and pomegranates [91]. Its consumption has been widely studied in relation to health promotion [92,93] due to their beneficial anti-inflammatory, anti-atherogenic, and antioxidant properties, among others [94,95]. The most abundant of these ET polyphenols are PC and EA [96]. However, PC and EA are not readily detected in human tissues or plasma after consumption of a high amount of pomegranate products [97]. It is now known that after the ingestion of pomegranate or pomegranate-based products, the ET polyphenols are poorly absorbed by the intestine because they are large (e.g., molecular weight of 1084.71 g/mol for PC) and hydrophobic, and the gut microbiota transforms them into potent metabolites called urolithins (Uro) [92,98] (Figure 1). Due to the poor bioavailability of PC and EA and extensive gut catabolism, it has been suggested that Uro, rather than EA and PC, are the actual bioactive molecules [99,100,101]. The circulation and distribution of PC, EA, and their metabolites have been studied in different tissues in human and animals, including pigs, sheep, birds, rodents, and insects [102,103]. In addition, Uro have been found to significantly accumulate in plasma and tissues [92,104,105]. As shown in Figure 1, PC from pomegranates and other sources is mainly hydrolyzed into EA in the acidic environment of the stomach. EA then undergoes a series of metabolic transformations by the gut microbiota to form Uro, with UroA and UroB being the two key final products.

4.2. Molecular Mechanisms Underlying the Beneficial Actions of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease

Some of the key anti-atherogenic actions of PC and its metabolites are indicated in Table 1 and summarized in Figure 2. These include antioxidant activities, effects on lipoprotein oxidation and metabolism, lipid accumulation and foam cell formation, and cytokine expression and inflammation together with impacts on disease-associated risk factors and the gut microbiota. These are addressed below in more detail.

4.2.1. Punicalagin and Metabolites as Antioxidants

Polyphenol compounds exert their antioxidant activity primarily via radical scavenging that, in vitro, involves the donation of an H atom or electron from their hydroxyl group to the free radical [117]. PC is known to have the most potent antioxidant activities when compared to other polyphenols [118]. The supplementation of pomegranate juice and pomegranate fruit extract rich in PC can significantly increase endothelial nitric oxide synthase (eNOS) activity, leading to the attenuation of pro-atherogenic-perturbed shear stress induced in vitro in human coronary ECs and in vivo in hypercholesterolemic mice [119]. Endothelial dysfunction due to high glucose is linked to the elevated generation of ROS and the treatment of human aortic ECs, as well as intact rat aortas with high glucose (30 mM) and EA, resulted in a significant decrease in ROS levels and improved the impaired vascular relaxation induced by the high glucose via the downregulation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) and the inhibition of extracellular signal-regulated kinase (ERK) 1/2-signaling pathways [120]. ROS can also cause cellular damage, particularly to DNA, RNA, proteins, and lipids, which can then lead to inflammation [121]. Uros-reduced ROS generation in both short- and long-term incubations by decreasing catalase, glutathione peroxidase, and superoxide dismutase enzymes in Caco-2 enterocytes, thereby preventing oxidative cellular damage [122]. In addition, the pre-treatment of human umbilical cord endothelial cells (HUVEC) with EA followed by exposure to oxLDL significantly attenuated ROS production, cytotoxicity, and apoptotic features by modulating eNOS and phosphoinositide 3-kinase pathways [123]. UroB showed antioxidant properties by reducing NADPH oxidase subunit expression and intracellular ROS production and inducing the antioxidant hememoxygenase-1 expression by nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element signaling in BV2 microglial cells [124]. Kelch-like ECH-associated protein 1 (Keap1) represses Nrf2 activity via multiple mechanisms; this can be interrupted by many proteins, including a ubiquitin-binding protein p62 [125]. UroB protected against myocardial ischemia/reperfusion injury via an increased accumulation of p62 and its subsequent interaction with Keap1, thereby resulting in protection against superoxide production and apoptotic cell death [126]. UroA also demonstrated anti-oxidative and neuroprotective actions in Alzheimer’s disease by inhibiting high glucose-induced amyloidogenesis produced by mitochondrial calcium dysregulation and mitochondrial ROS accumulation [127].

4.2.2. Effects of Punicalagin and Its Metabolites on Lipoprotein Metabolism and Lipid Homeostasis

Lipoproteins, including LDL, HDL, and their oxidatively modified forms, play vital roles in cholesterol metabolism and associated disorders [22]. In addition, foam cell formation during atherosclerosis is controlled by the uptake, intracellular metabolism, and efflux of cholesterol in macrophages and VSMC [12]. Furthermore, lipid metabolism in other tissues (e.g., liver, adipose tissue) impacts lipid homeostasis in atherosclerotic plaques [12]. Polyphenols such as PC have many protective actions against lipid and lipoprotein homeostasis, as well as processes regulated by their dysfunction [128,129]. Thus, a randomized, double-blinded, placebo-controlled, crossover trial performed on 67 healthy adults for 20 weeks to evaluate the effects of hydroxytryrosol and PC on early atherosclerosis-associated markers showed that supplementation with these two polyphenols exerted anti-atherosclerotic effects by improving blood pressure, endothelial function, and decreasing the levels of circulating oxLDL [88]. In an in vivo study, Wistar rats were fed with a high-cholesterol diet supplemented with Vitamin D3 and subjected to the balloon injury of the aorta. UroA (3 mg/kg/day) produced a significant improvement in the plasma lipid profile and Angiotensin II levels together with aortic lesions compared with the control group [111]. UroB also decreased lipid plaque deposition in Apolipoprotein E-deficient (ApoE−/−) mice and enhanced macrophage cholesterol efflux through the induced expression of SR-BI and ABCA1 [107]. The combination of pomegranate and dates in ApoE−/− mice reduced plasma cholesterol and TG levels associated with increased paraoxonase activity and lipid peroxide content in the aorta and produced significant decrease in oxidative stress, cholesterol content, and LDL uptake in peritoneal macrophages [130]. In in vitro studies, the treatment of murine J774A.1 macrophages with PC-enhanced statin-mediated cholesterol biosynthesis and protected against foam cell formation [110,130]. EA also decreased the oxLDL-mediated foam cell formation in J774A.1 macrophages and enhanced cholesterol efflux from foam cells [109].
The impact of PC, EA, and its metabolites on lipid metabolism is not just restricted to macrophages but extends to other cellular systems. Thus, UroA, UroB, and UroC reduced TG accumulation and fatty acid oxidation in adipocytes and hepatocytes [131]. EA and UroA attenuated lipid accumulation in 3T3–L1 adipocytes through the regulation of glucose Transporter Type 4 and adiponectin [132]. In streptozotocin-induced Type 1 diabetes mellitus in rats, EA prevented hepatic lipid accumulation and reduced both hepatic and plasma levels of TG, cholesterol, and FFAs by activating AMP-activated protein kinase [133]. Furthermore, EA decreased plasma cholesterol and TG levels and increased fecal bile acid excretion in hamsters. This was associated with an increased expression of liver X receptor-α, peroxisome proliferator-activated receptor -γ, and -α, together with their downstream target gene ABCA1 [134].

4.2.3. Effects of PC and Its Metabolites on Inflammation and Expression of Cytokines

Anti-inflammatory effects of PC and its metabolites are widely highlighted in the literature [135,136,137]. Thus, UroA significantly attenuated the production of pro-inflammatory mediators in lipopolysaccharide (LPS)-stimulated RAW264 macrophages, leading to the inhibition of Akt and c-Jun N-terminal kinase phosphorylation and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and activator protein-1 activation [138]. UroA also inhibited TNF-α-induced expression of both MCP-1 and IL-8 in human aortic ECs together with their migration and adhesion to monocytes [94]. In addition, an in vitro study in THP-1 macrophages, in which IFN-γ was used to induce the expression of several pro-inflammatory genes, in particular ICAM-1 and MCP-1, PC significantly inhibited their expression together with MCP-1-induced monocytic migration [139]. UroA also reduced the expression of various inflammatory factors in response to the oxLDL stimulation of human artery ECs, such as MCP-1, ICAM-1, TNF-α, and IL-6. Consequently, it attenuated the adhesion of monocytes to these cells [94,116]. In another study, in human placenta, visceral adipose tissue and subcutaneous adipose tissue explants, treatment with PC and curcumin significantly suppressed the TNF-α-induced expression of chemokines [C-C motif ligand (CCL)2-5, C-X-C motif ligand (CXCL)1, CXCL5, CXCL8] and pro-inflammatory cytokines (IL-1α, IL-1β, IL-6) [140]. EA also inhibited oxLDL- and IL-1 β-mediated activation of NF-κB, as well as downstream targets, such as cytokines and adhesion proteins, in HUVEC [114,141]. UroB also inhibited the production of pro-inflammatory cytokines and increased an anti-inflammatory cytokine IL-10 in LPS-stimulated BV2 microglial cells [124].
As detailed above, in vivo, UroB attenuated atherosclerosis, an inflammatory disorder, in ApoE−/− mice [107]. In male Balb/c mice fed a HFD for 12 weeks and given PC subcutaneously in the last four weeks, there was a significant improvement in HDL anti-inflammatory properties and other anti-inflammatory parameters compared to the control [137]. The oral supplementation of EA also downregulated the expression of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α in isoproterenol-treated rats and protected against cardiac damage [142]. The effect of UroB after MI was investigated in vivo using adult male Sprague Dawley rats and revealed cardioprotective effects through the control of inflammation and cardiac fibrosis [115].

4.2.4. Effects of PC and Its Metabolites on Other Pathologies That Impact Atherosclerosis

Other inflammatory disorders, such as rheumatoid arthritis, asthma, Type 2 diabetes, and NAFLD, also impact atherosclerosis [25]. PC and its metabolites also affect these disorders and, therefore, atherosclerosis; this is illustrated effectively in Figure 3 in relation to NAFLD. Thus, in an in vivo study, EA improved hepatic steatosis and the plasma lipid profile in the KK-A(y) mice that were fed HFD as a model for obese Type 2 diabetes [143]. PC (20 mg/kg body weight/day) protected against HFD and streptozotocin-induced diabetic liver injury in C57BL/6 mice via the activation of antioxidant enzymes and upregulation of mitophagy [144]. UroA also decreased ROS levels in HepG2 cells, together with the expression of NF-κB p65 and other inflammatory markers, and improved antioxidant activities [145]. It has been reported that UroA, UroC, and UroD also increase fatty acid oxidation and attenuate TG accumulation in hepatocytes and adipocytes [131]. In relation to other inflammatory disorders, EA was found to inhibit IL-1β-induced activation of NF-κB signaling and several downstream genes in human chondrocytes and protected in a surgical DMM (destabilization of the medical meniscus) model of osteoarthritis [146]. In an ovalalbumin-induced mouse asthma model, EA also inhibited NF-κB activation and the development of airway hyper responsiveness (e.g., lung eosinophilic inflammation and goblet cell hyperplasia) [147].

4.2.5. The Impact of PC and Its Metabolites on the Gut Microbiota

Polyphenols can influence the gut microbiota in a manner that encourages the growth of beneficial bacteria while inhibiting the growth of harmful bacteria [148]. In addition, the microbiome can influence polyphenols to become more bioavailable via metabolism into new metabolites [149]. For example, the gut bacteria Gordonibacter pamelaeae and Gordonibacter urolithinfaciens have shown the potential to biotransform EA to urolithins [150]. PC can alleviate insulin resistance by regulating gut microbiota and autophagy [151]. Some of the actions of PC and its metabolites may potentially be mediated via the modulation of the gut microbiota. Thus, UroA and UroB possess anti-obesity properties in a HFD-induced rat model of obesity via modulation of the gut microbiota [152]. Polyphenols often cause the production of short chain fatty acids [153,154], such as propionate and butyrate, which then prevents ACVD [155,156].

5. Conclusions

PC and its metabolites have protective actions on atherosclerosis and other inflammatory disorders via multiple mechanisms, including antioxidant and anti-inflammatory properties, together with the modulation of the gut microbiota. However, studies on the role of PC and its metabolites in mouse models of atherosclerosis have been limited to PC and UroB [107,130]. In addition, these studies have been rather restricted to monitoring only some parameters (e.g., plaque lipid content). More detailed studies are required that investigate the effects of PC and its metabolites on plaque burden, lipid content, and cellularity (e.g., levels of macrophages, T-cells and various subtypes, other immune cells, smooth muscle cells etc), using a combination of histological and immunohistological analyses, similar to those carried out on other nutraceuticals [157]. Such studies can provide key information on whether PC and its metabolites can dampen plaque inflammation and stabilize existing plaques, thereby informing on mechanisms of actions and therapeutic potential. The studies in such models can be extended to plasma lipid profile, immune cell profile in peripheral blood, bone marrow, spleen, and thymus together with other tissues (e.g., liver in relation to NAFLD). Gene expression analysis using arrays, RNA-sequencing (RNA-seq), and single-cell RNA-seq on the aorta and the liver [157,158,159] can provide insights into pathways regulated by PC and its metabolites in atherosclerosis and NAFLD, and whether there are common genes/pathways regulated by different agents. The identification of key pathways can also form the foundation of drug discovery programs to screen for potent agonists. More mechanistic insights can also be obtained using the full range of in vitro assays on all the different cell types present in atherosclerotic plaques, including macrophages, ECs, and SMCs [160]. Such assays include cell proliferation and migration, foam cell formation, inflammasome activation, endothelial cell dysfunction, and SMC phenotypic shift. The in vitro and in vivo assays should employ the full range of physiological doses and can be extended further to investigate whether PC and its metabolites can also cause a regression of existing atherosclerotic plaques in mouse model systems [159]. Combining agents may provide additional insights on the existence of any synergistic or antagonistic actions [161]. Such studies can then form the foundations for large clinical trials; initial studies in humans have already shown promise in relation to improved mitochondrial and cellular health [112]. Given the existence of co-morbidities, studies on PC and its metabolites should ultimately be extended beyond atherosclerosis and NAFLD to include neurological disorders, metabolic syndrome, diabetes, and obesity.

Funding

Sultanate of Oman and Kingdom of Saudi Arabia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

S.A. and F.A. received studentships from Sultanate of Oman and Kingdom of Saudi Arabia, respectively.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABC: ATP-binding cassette transporter; ACAT1, acyl-CoA acyltransferase-1; Apo, apolipoprotein; ApoE−/−, apolipoprotein E deficient; AVCD, atherosclerotic cardiovascular disease; CCL, C-C motif ligand; CD36, cluster of differentiation 36; CE, cholesteryl ester; CETP, cholesteryl ester transfer protein; CVD, cardiovascular disease; CXC, C-X-C motif ligand; EA, ellagic acids; EC, endothelial cells; ECM, extracellular matrix; eNOS, endothelial nitric oxide synthase; ERK, extracellular signal-regulated kinase; ET, elagitannins; FFA, free fatty acids; HDL, high-density lipoprotein; HUVEC, human umbilical cord endothelial cells; ICAM-1, intercellular adhesion molecule-1; IFN, interferon; IL, interleukin; Keap1, Kelch-like ECH-associated protein 1; LDL, low-density lipoprotein; LDLr, LDL receptor; LOX-1, lectin-like oxLDL receptot-1; LPS, lipopolysaccharide; MCP-1, monocyte chemotactic protein-1; MI, myocardial infarction; NADPH, nicotinamide adenine dinucleotide phosphate; Nox4, NADPH oxidase 4; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2, nuclear factor erythroid 2-related factor 2; oxLDL, oxidized LDL; PC, punicalagin; RCT, reverse cholesterol transport; ROS, reactive oxygen species; RNA-seq, RNA-sequencing; RNS, reactive nitrogen species; SMC, smooth muscle cells; SR-A1, scavenger receptor type 1; SR-B1, scavenger receptor class B type 1; TFEB, transcription factor EB; TG, triacylglycerol; TNF, tumour necrosis factor; Uro, urolithins; VCAM-1, vascular cell adhesion molecule-1; VSMC, vascular smooth muscle cells.

References

  1. Chan, Y.H.; Ramji, D.P. Atherosclerosis: Pathogenesis and key cellular processes, current and emerging therapies, key challenges, and future research directions. Methods Mol. Biol. 2022, 2419, 3–19. [Google Scholar]
  2. Bhatnagar, P.; Wickramasinghe, K.; Williams, J.; Rayner, M.; Townsend, N. The epidemiology of cardiovascular disease in the UK 2014. Heart 2015, 101, 1182–1189. [Google Scholar] [CrossRef] [PubMed]
  3. Wilson, L.; Bhatnagar, P.; Townsend, N. Comparing trends in mortality from cardiovascular disease and cancer in the United Kingdom, 1983–2013: Joinpoint regression analysis. Popul. Health Metr. 2017, 15, 23. [Google Scholar] [CrossRef]
  4. Spirig, R.; Tsui, J.; Shaw, S. The emerging role of TLR and innate immunity in cardiovascular disease. Cardiol. Res. Pract. 2012, 2012, 181394. [Google Scholar] [CrossRef]
  5. Libby, P.; Mallat, Z.; Weyand, C. Immune and inflammatory mechanisms mediate cardiovascular diseases from head to toe. Cardiovasc. Res. 2021, 117, 2503–2505. [Google Scholar] [CrossRef] [PubMed]
  6. Chan, Y.H.; Ramji, D.P. Key roles of inflammation in atherosclerosis: Mediators involved in orchestrating the inflammatory response and its resolution in the disease along with therapeutic avenues targeting inflammation. Methods Mol. Biol. 2022, 2419, 21–37. [Google Scholar] [PubMed]
  7. Virani, S.S.; Alonso, A.; Aparicio, H.J.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Cheng, S.; Delling, F.N. Heart disease and stroke statistics—2021 update: A report from the American Heart Association. Circulation 2021, 143, e254–e743. [Google Scholar] [CrossRef] [PubMed]
  8. Ramji, D.P.; Davies, T.S. Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev. 2015, 26, 673–685. [Google Scholar] [CrossRef]
  9. Chan, Y.-H.; Ramji, D.P. A perspective on targeting inflammation and cytokine actions in atherosclerosis. Future Med. Chem. 2020, 12, 613–626. [Google Scholar] [CrossRef]
  10. van der Vorst, E.P.C.; Döring, Y.; Weber, C. Chemokines and their receptors in atherosclerosis. J. Mol. Med. 2015, 93, 963–971. [Google Scholar] [CrossRef]
  11. Reglero-Real, N.; Colom, B.; Bodkin, J.V.; Nourshargh, S. Endothelial cell junctional adhesion molecules: Role and regulation of expression in inflammation. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 2048–2057. [Google Scholar] [CrossRef]
  12. McLaren, J.E.; Michael, D.R.; Ashlin, T.G.; Ramji, D.P. Cytokines, macrophage lipid metabolism and foam cells: Implications for cardiovascular disease therapy. Prog. Lipid Res. 2011, 50, 331–347. [Google Scholar] [CrossRef]
  13. Martinet, W.; Coornaert, I.; Puylaert, P.; De Meyer, G.R.Y. Macrophage death as a pharmacological target in atherosclerosis. Front. Pharmacol. 2019, 10, 306. [Google Scholar] [CrossRef]
  14. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  15. Chan, Y.H.; Ramji, D.P. Probing inflammasome activation in atherosclerosis. Methods Mol. Biol. 2022, 2419, 313–331. [Google Scholar] [PubMed]
  16. Kumar, V.; Prabhu, S.D.; Bansal, S.S. CD4+ T-lymphocytes exhibit biphasic kinetics post-myocardial infarction. Front. Cardiovasc. Med. 2022, 9, 992653. [Google Scholar] [CrossRef]
  17. Kumar, V.; Rosenzweig, R.; Asalla, A.; Nehra, S.; Prabhu, S.D.; Bansal, S.S. TNFR1 contributes to activation-induced cell death of pathological CD4+ T lymphocytes during ischemic heart failure. JACC Basic Transl. Sci. 2022, 7, 1038–1049. [Google Scholar] [CrossRef] [PubMed]
  18. Lu, Y.; Xia, N.; Cheng, X. Regulatory T cells in chronic heart failure. Front. Immunol. 2021, 12, 732794. [Google Scholar] [CrossRef]
  19. Doran, A.C.; Yurdagul, A.; Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 2020, 20, 254–267. [Google Scholar] [CrossRef] [PubMed]
  20. Van Vré, E.A.; Ait-Oufella, H.; Tedgui, A.; Mallat, Z. Apoptotic cell death and efferocytosis in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 887–893. [Google Scholar] [CrossRef]
  21. Liu, X.; Ni, M.; Ma, L.; Yang, J.; Wang, L.; Liu, F.; Dong, M.; Yang, X.; Zhang, M.; Lu, H. Targeting blood thrombogenicity precipitates atherothrombotic events in a mouse model of plaque destabilization. Sci. Rep. 2015, 5, 10225. [Google Scholar] [CrossRef] [PubMed]
  22. Buckley, M.L.; Ramji, D.P. The influence of dysfunctional signaling and lipid homeostasis in mediating the inflammatory responses during atherosclerosis. Biochim. Biophys. Acta 2015, 1852, 1498–1510. [Google Scholar] [CrossRef]
  23. O’Morain, V.L.; Ramji, D.P. The potential of probiotics in the prevention and treatment of atherosclerosis. Mol. Nutr. Food Res. 2020, 64, e1900797. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, D.; Mi, Z.; Peng, J.; Yang, T.; Han, Y.; Zhai, Y.; Song, C.; Teng, X.; Sun, W.; Guo, J.; et al. Non-alcoholic fatty liver disease as an emerging risk factor and potential intervention target for atherosclerotic cardiovascular diseases. J. Cardiovasc. Pharmacol. 2023. [Google Scholar] [CrossRef] [PubMed]
  25. Hassen, G.; Singh, A.; Belete, G.; Jain, N.; De la Hoz, I.; Camacho-Leon, G.P.; Dargie, N.K.; Carrera, K.G.; Alemu, T.; Jhaveri, S.; et al. Nonalcoholic fatty liver disease: An emerging modern-day risk factor for cardiovascular disease. Cureus 2022, 14, e25495. [Google Scholar] [CrossRef]
  26. Zhang, L.; She, Z.G.; Li, H.; Zhang, X.J. Non-alcoholic fatty liver disease: A metabolic burden promoting atherosclerosis. Clin. Sci. 2020, 134, 1775–1799. [Google Scholar] [CrossRef]
  27. Pei, K.; Gui, T.; Kan, D.; Feng, H.; Jin, Y.; Yang, Y.; Zhang, Q.; Du, Z.; Gai, Z.; Wu, J. An overview of lipid metabolism and nonalcoholic fatty liver disease. BioMed Res. Int. 2020, 2020, 4020249. [Google Scholar] [CrossRef] [PubMed]
  28. Li, H.; Yu, X.-H.; Ou, X.; Ouyang, X.-P.; Tang, C.-K. Hepatic cholesterol transport and its role in non-alcoholic fatty liver disease and atherosclerosis. Prog. Lipid Res. 2021, 83, 101109. [Google Scholar]
  29. Stols-Gonçalves, D.; Hovingh, G.K.; Nieuwdorp, M.; Holleboom, A.G. NAFLD and atherosclerosis: Two sides of the same dysmetabolic coin? Trends Endocrinol. Metab. 2019, 30, 891–902. [Google Scholar] [CrossRef]
  30. Li, W.; Liu, J.; Cai, J.; Zhang, X.-J.; Zhang, P.; She, Z.-G.; Chen, S.; Li, H. NAFLD as a continuous driver in the whole spectrum of vascular disease. J. Mol. Cell. Cardiol. 2022, 163, 118–132. [Google Scholar] [CrossRef]
  31. Wang, Z.; Ye, M.; Zhang, X.-J.; Zhang, P.; Cai, J.; Li, H.; She, Z.-G. Impact of NAFLD and its pharmacotherapy on lipid profile and CVD. Atherosclerosis 2022, 355, 30–44. [Google Scholar] [CrossRef]
  32. Weidinger, A.; Kozlov, A.V. Biological activities of reactive oxygen and nitrogen species: Oxidative stress versus signal transduction. Biomolecules 2015, 5, 472–484. [Google Scholar] [CrossRef] [PubMed]
  33. Cojocaru, K.A.; Luchian, I.; Goriuc, A.; Antoci, L.M.; Ciobanu, C.G.; Popescu, R.; Vlad, C.E.; Blaj, M.; Foia, L.G. Mitochondrial dysfunction, oxidative stress, and therapeutic strategies in diabetes, obesity, and cardiovascular disease. Antioxidants 2023, 12, 658. [Google Scholar] [CrossRef]
  34. Förstermann, U.; Xia, N.; Li, H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef] [PubMed]
  35. Dikalov, S.; Itani, H.; Richmond, B.; Arslanbaeva, L.; Vergeade, A.; Rahman, S.M.J.; Boutaud, O.; Blackwell, T.; Massion, P.P.; Harrison, D.G. Tobacco smoking induces cardiovascular mitochondrial oxidative stress, promotes endothelial dysfunction, and enhances hypertension. Am. J. Physiol.-Heart Circ. Physiol. 2019, 316, H639–H646. [Google Scholar] [CrossRef] [PubMed]
  36. Burtenshaw, D.; Kitching, M.; Redmond, E.M.; Megson, I.L.; Cahill, P.A. Reactive oxygen species (ROS), intimal thickening, and subclinical atherosclerotic disease. Front. Cardiovasc. Med. 2019, 6, 89. [Google Scholar] [CrossRef] [PubMed]
  37. Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019, 25, 101084. [Google Scholar] [CrossRef]
  38. Hume, D.A.; Irvine, K.M.; Pridans, C. The mononuclear phagocyte system: The relationship between monocytes and macrophages. Trends Immunol. 2019, 40, 98–112. [Google Scholar] [CrossRef]
  39. Fujiwara, N.; Kobayashi, K. Macrophages in inflammation. Curr. Drug Target. Inflamm. Allergy 2005, 4, 281–286. [Google Scholar] [CrossRef]
  40. Regoes, R.R.; McLaren, P.J.; Battegay, M.; Bernasconi, E.; Calmy, A.; Günthard, H.F.; Hoffmann, M.; Rauch, A.; Telenti, A.; Fellay, J.; et al. Disentangling human tolerance and resistance against HIV. PLoS Biol. 2014, 12, e1001951. [Google Scholar] [CrossRef]
  41. Bloomer, S.A.; Moyer, E.D.; Brown, K.E.; Kregel, K.C. Aging results in accumulation of M1 and M2 hepatic macrophages and a differential response to gadolinium chloride. Histochem. Cell Biol. 2020, 153, 37–48. [Google Scholar] [CrossRef] [PubMed]
  42. Flynn, M.C.; Pernes, G.; Lee, M.K.S.; Murphy, A.J.; Nagareddy, P.R. Monocytes, macrophages and metabolic disease in atherosclerosis. Front. Pharmacol. 2019, 10, 666. [Google Scholar] [CrossRef] [PubMed]
  43. Prenen, H.; Mazzone, M. Tumor-associated macrophages: A short compendium. Cell. Mol. Life Sci. 2019, 76, 1447–1458. [Google Scholar] [CrossRef]
  44. Moore, K.J.; Sheedy, F.J.; Fisher, E.A. Macrophages in atherosclerosis: A dynamic balance. Nat. Rev. Immunol. 2013, 13, 709–721. [Google Scholar] [CrossRef]
  45. Moss, J.W.; Ramji, D.P. Cytokines: Roles in atherosclerosis disease progression and potential therapeutic targets. Future Med. Chem. 2016, 8, 1317–1330. [Google Scholar] [CrossRef]
  46. Fatkhullina, A.R.; Peshkova, I.O.; Koltsova, E.K. The role of cytokines in the development of atherosclerosis. Biochemistry 2016, 81, 1358–1370. [Google Scholar] [CrossRef]
  47. Ait-Oufella, H.; Taleb, S.; Mallat, Z.; Tedgui, A. Recent advances on the role of cytokines in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 969–979. [Google Scholar] [CrossRef]
  48. Theofilis, P.; Sagris, M.; Oikonomou, E.; Antonopoulos, A.S.; Siasos, G.; Tsioufis, C.; Tousoulis, D. Inflammatory mechanisms contributing to endothelial dysfunction. Biomedicines 2021, 9, 781. [Google Scholar] [CrossRef]
  49. Basatemur, G.L.; Jørgensen, H.F.; Clarke, M.C.H.; Bennett, M.R.; Mallat, Z. Vascular smooth muscle cells in atherosclerosis. Nat. Rev. Cardiol. 2019, 16, 727–744. [Google Scholar] [CrossRef] [PubMed]
  50. Remmerie, A.; Scott, C.L. Macrophages and lipid metabolism. Cell. Immunol. 2018, 330, 27–42. [Google Scholar] [CrossRef]
  51. Fennelly, C.; Amaravadi, R.K. Lysosomal biology in cancer. Methods Mol. Biol. 2017, 1594, 293–308. [Google Scholar] [PubMed]
  52. Marques, A.R.A.; Ramos, C.; Machado-Oliveira, G.; Vieira, O.V. Lysosome (dys)function in atherosclerosis-A big weight on the shoulders of a small organelle. Front. Cell. Dev. Biol. 2021, 9, 658995. [Google Scholar] [CrossRef] [PubMed]
  53. Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef]
  54. Sergin, I.; Evans, T.D.; Zhang, X.; Bhattacharya, S.; Stokes, C.J.; Song, E.; Ali, S.; Dehestani, B.; Holloway, K.B.; Micevych, P.S. Exploiting macrophage autophagy-lysosomal biogenesis as a therapy for atherosclerosis. Nat. Commun. 2017, 8, 15750. [Google Scholar] [CrossRef]
  55. Javaheri, A.; Bajpai, G.; Picataggi, A.; Mani, S.; Foroughi, L.; Evie, H.; Kovacs, A.; Weinheimer, C.J.; Hyrc, K.; Xiao, Q. TFEB activation in macrophages attenuates postmyocardial infarction ventricular dysfunction independently of ATG5-mediated autophagy. JCI Insight 2019, 4, e127312. [Google Scholar] [CrossRef] [PubMed]
  56. Haas, M.J.; Feng, V.; Gonzales, K.; Bikkina, P.; Angelica Landicho, M.; Mooradian, A.D. Transcription factor EB protects against endoplasmic reticulum stress in human coronary artery endothelial cells. Eur. J. Pharmacol. 2022, 933, 175274. [Google Scholar] [CrossRef]
  57. Li, M.; Wang, Z.; Wang, P.; Li, H.; Yang, L. TFEB: A emerging regulator in lipid homeostasis for atherosclerosis. Front. Physiol. 2021, 12, 639920. [Google Scholar] [CrossRef]
  58. Lu, H.; Sun, J.; Hamblin, M.H.; Chen, Y.E.; Fan, Y. Transcription factor EB regulates cardiovascular homeostasis. EBioMedicine 2021, 63, 103207. [Google Scholar] [CrossRef] [PubMed]
  59. Evans, T.D.; Jeong, S.J.; Zhang, X.; Sergin, I.; Razani, B. TFEB and trehalose drive the macrophage autophagy-lysosome system to protect against atherosclerosis. Autophagy 2018, 14, 724–726. [Google Scholar] [CrossRef]
  60. Wong, W. Protected from atherosclerosis by TFEB. Science 2017, 355, 490. [Google Scholar] [CrossRef]
  61. Lu, H.; Fan, Y.; Qiao, C.; Liang, W.; Hu, W.; Zhu, T.; Zhang, J.; Chen, Y.E. TFEB inhibits endothelial cell inflammation and reduces atherosclerosis. Sci. Signal. 2017, 10, 464. [Google Scholar] [CrossRef] [PubMed]
  62. Emanuel, R.; Sergin, I.; Bhattacharya, S.; Turner, J.N.; Epelman, S.; Settembre, C.; Diwan, A.; Ballabio, A.; Razani, B. Induction of lysosomal biogenesis in atherosclerotic macrophages can rescue lipid-induced lysosomal dysfunction and downstream sequelae. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1942–1952. [Google Scholar] [CrossRef] [PubMed]
  63. Griffin, E.E.; Ullery, J.C.; Cox, B.E.; Jerome, W.G. Aggregated LDL and lipid dispersions induce lysosomal cholesteryl ester accumulation in macrophage foam cells. J. Lipid Res. 2005, 46, 2052–2060. [Google Scholar] [CrossRef]
  64. Yuan, Y.; Chen, Y.; Peng, T.; Li, L.; Zhu, W.; Liu, F.; Liu, S.; An, X.; Luo, R.; Cheng, J. Mitochondrial ROS-induced lysosomal dysfunction impairs autophagic flux and contributes to M1 macrophage polarization in a diabetic condition. Clin. Sci. 2019, 133, 1759–1777. [Google Scholar] [CrossRef]
  65. Hussain, M.M. Intestinal lipid absorption and lipoprotein formation. Curr. Opin. Lipidol. 2014, 25, 200. [Google Scholar] [CrossRef] [PubMed]
  66. Ouimet, M.; Barrett, T.J.; Fisher, E.A. HDL and reverse cholesterol transport: Basic mechanisms and their roles in vascular health and disease. Circ. Res. 2019, 124, 1505–1518. [Google Scholar] [CrossRef]
  67. Chen, W.; Wang, N.; Tall, A.R. A PEST deletion mutant of ABCA1 shows impaired internalization and defective cholesterol efflux from late endosomes. J. Biol. Chem. 2005, 280, 29277–29281. [Google Scholar] [CrossRef]
  68. Tarling, E.J.; Edwards, P.A. ATP binding cassette transporter G1 (ABCG1) is an intracellular sterol transporter. Proc. Natl. Acad. Sci. USA 2011, 108, 19719–19724. [Google Scholar] [CrossRef]
  69. Liu, X.; Xiong, S.L.; Yi, G.-H. ABCA1, ABCG1, and SR-BI: Transit of HDL-associated sphingosine-1-phosphate. Clin. Chim. Acta 2012, 413, 384–390. [Google Scholar] [CrossRef] [PubMed]
  70. Vitali, C.; Cuchel, M. Controversial role of lecithin:cholesterol acyltransferase in the development of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 377–379. [Google Scholar]
  71. Trajkovska, K.T.; Topuzovska, S. High-density lipoprotein metabolism and reverse cholesterol transport: Strategies for raising HDL cholesterol. Anatol. J. Cardiol. 2017, 18, 149. [Google Scholar] [CrossRef] [PubMed]
  72. Oliveira, H.C.F.; Raposo, H.F. Lipid Transfer in Lipoprotein Metabolism and Cardiovascular Disease; Springer: Berlin/Heidelberg, Germany, 2020; pp. 15–25. [Google Scholar]
  73. Alphonse, P.A.S.; Jones, P.J.H. Revisiting human cholesterol synthesis and absorption: The reciprocity paradigm and its key regulators. Lipids 2016, 51, 519–536. [Google Scholar] [CrossRef]
  74. Koushki, K.; Shahbaz, S.K.; Mashayekhi, K.; Sadeghi, M.; Zayeri, Z.D.; Taba, M.Y.; Banach, M.; Al-Rasadi, K.; Johnston, T.P.; Sahebkar, A. Anti-inflammatory action of statins in cardiovascular disease: The role of inflammasome and toll-like receptor pathways. Clin. Rev. Allergy Immunol. 2021, 60, 175–199. [Google Scholar] [PubMed]
  75. Almeida, S.O.; Budoff, M. Effect of statins on atherosclerotic plaque. Trends Cardiovasc. Med. 2019, 29, 451–455. [Google Scholar] [CrossRef]
  76. Pinal-Fernandez, I.; Casal-Dominguez, M.; Mammen, A.L. Statins: Pros and cons. Med. Clín. 2018, 150, 398–402. [Google Scholar]
  77. Ladeiras-Lopes, R.; Agewall, S.; Tawakol, A.; Staels, B.; Stein, E.; Mentz, R.J.; Leite-Moreira, A.; Zannad, F.; Koenig, W. Atherosclerosis: Recent trials, new targets and future directions. Int. J. Cardiol. 2015, 192, 72–81. [Google Scholar] [CrossRef] [PubMed]
  78. Cicero, A.F.G.; Caliceti, C.; Fogacci, F.; Giovannini, M.; Calabria, D.; Colletti, A.; Veronesi, M.; Roda, A.; Borghi, C. Effect of apple polyphenols on vascular oxidative stress and endothelium function: A translational study. Mol. Nutr. Food Res. 2017, 61, 1700373. [Google Scholar] [CrossRef]
  79. Szulińska, M.; Skrypnik, D.; Michałowska, J.; Bogdański, P. Non-pharmacological modification of endothelial function: An important lesson for clinical practice. Adv. Hyg. Exp. Med. (PHMD) 2018, 72, 89–100. [Google Scholar] [CrossRef]
  80. Moss, J.W.E.; Williams, J.O.; Ramji, D.P. Nutraceuticals as therapeutic agents for atherosclerosis. Biochim. Biophys. Acta 2018, 1864, 1562–1572. [Google Scholar] [CrossRef]
  81. Moss, J.W.E.; Ramji, D.P. Nutraceutical therapies for atherosclerosis. Nat. Rev. Cardiol. 2016, 13, 513–532. [Google Scholar] [CrossRef]
  82. Tresserra-Rimbau, A.; Rimm, E.B.; Medina-Remón, A.; Martínez-González, M.A.; De la Torre, R.; Corella, D.; Salas-Salvadó, J.; Gómez-Gracia, E.; Lapetra, J.; Arós, F. Inverse association between habitual polyphenol intake and incidence of cardiovascular events in the PREDIMED study. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 639–647. [Google Scholar] [CrossRef] [PubMed]
  83. Ed Nignpense, B.; Chinkwo, K.A.; Blanchard, C.L.; Santhakumar, A.B. Polyphenols: Modulators of platelet function and platelet microparticle generation? Int. J. Mol. Sci. 2019, 21, 146. [Google Scholar] [CrossRef]
  84. Lockyer, S.; Rowland, I.; Spencer, J.P.E.; Yaqoob, P.; Stonehouse, W. Impact of phenolic-rich olive leaf extract on blood pressure, plasma lipids and inflammatory markers: A randomised controlled trial. Eur. J. Nutr. 2017, 56, 1421–1432. [Google Scholar] [CrossRef]
  85. Yamagata, K. Polyphenols regulate endothelial functions and reduce the risk of cardiovascular disease. Curr. Pharm. Des. 2019, 25, 2443–2458. [Google Scholar]
  86. Aviram, M.; Rosenblat, M. Pomegranate protection against cardiovascular diseases. Evid. Based Complement. Altern. Med. 2012, 2012, 382763. [Google Scholar] [CrossRef] [PubMed]
  87. Benchagra, L.; Hajjaji, A.; Ramchoun, M.; Khalil, A.; Berrougui, H. Beneficial effects of pomegranate fruit consumption in cardiovascular diseases prevention. J. Nutr. Ther. 2019, 7, 84–94. [Google Scholar] [CrossRef]
  88. Quirós-Fernández, R.; López-Plaza, B.; Bermejo, L.M.; Palma-Milla, S.; Gómez-Candela, C. Supplementation with hydroxytyrosol and punicalagin improves early atherosclerosis markers Involved in the asymptomatic phase of atherosclerosis in the adult population: A randomized, placebo-controlled, crossover trial. Nutrients 2019, 11, 640. [Google Scholar] [CrossRef]
  89. Mathon, C.; Chater, J.M.; Green, A.; Merhaut, D.J.; Mauk, P.A.; Preece, J.E.; Larive, C.K. Quantification of punicalagins in commercial preparations and pomegranate cultivars, by liquid chromatography–mass spectrometry. J. Sci. Food Agric. 2019, 99, 4036–4042. [Google Scholar] [CrossRef]
  90. Seeram, N.P.; Adams, L.S.; Henning, S.M.; Niu, Y.; Zhang, Y.; Nair, M.G.; Heber, D. In vitro antiproliferative, apoptotic and antioxidant activities of punicalagin, ellagic acid and a total pomegranate tannin extract are enhanced in combination with other polyphenols as found in pomegranate juice. J. Nutr. Bioch. 2005, 16, 360–367. [Google Scholar] [CrossRef] [PubMed]
  91. Okuda, T.; Yoshida, T.; Hatano, T.; Ito, H. Chemistry and Biology of Ellagitannins: An Underestimated Class of Bioactive Plant Polyphenols; World Scientific Press: Singapore, 2009; pp. 1–54. [Google Scholar]
  92. Cerdá, B.; Espín, J.C.; Parra, S.; Martínez, P.; Tomás-Barberán, F.A. The potent in vitro antioxidant ellagitannins from pomegranate juice are metabolised into bioavailable but poor antioxidant hydroxy–6H–dibenzopyran–6–one derivatives by the colonic microflora of healthy humans. Eur. J. Nutr. 2004, 43, 205–220. [Google Scholar] [CrossRef]
  93. Ismail, T.; Calcabrini, C.; Diaz, A.R.; Fimognari, C.; Turrini, E.; Catanzaro, E.; Akhtar, S.; Sestili, P. Ellagitannins in cancer chemoprevention and therapy. Toxins 2016, 8, 151. [Google Scholar] [CrossRef] [PubMed]
  94. Espın, J.C.; Garcıa-Conesa, M.-T. Ellagitannin metabolites, urolithin A glucuronide and its aglycone urolithin A, ameliorate TNF--induced inflammation and associated molecular markers in human aortic endothelial cells. Mol. Nutr. Food Res. 2012, 56, 784–796. [Google Scholar]
  95. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouysegu, L. Plant polyphenols: Chemical properties, biological activities, and synthesis. Angew. Chem. Int. Ed. 2011, 50, 586–621. [Google Scholar] [CrossRef] [PubMed]
  96. Gil, M.I.; Tomás-Barberán, F.A.; Hess-Pierce, B.; Holcroft, D.M.; Kader, A.A. Antioxidant activity of pomegranate juice and its relationship with phenolic composition and processing. J. Agric. Food Chem. 2000, 48, 4581–4589. [Google Scholar] [CrossRef]
  97. Mertens-Talcott, S.U.; Jilma-Stohlawetz, P.; Rios, J.; Hingorani, L.; Derendorf, H. Absorption, metabolism, and antioxidant effects of pomegranate (Punica granatum L.) polyphenols after ingestion of a standardized extract in healthy human volunteers. J. Agric. Food Chem. 2006, 54, 8956–8961. [Google Scholar] [CrossRef]
  98. Garcia-Villalba, R.; Vissenaekens, H.; Pitart, J.; Romo-Vaquero, M.; Espín, J.C.; Grootaert, C.; Selma, M.V.; Raes, K.; Smagghe, G.; Possemiers, S. Gastrointestinal simulation model TWIN-SHIME shows differences between human urolithin-metabotypes in gut microbiota composition, pomegranate polyphenol metabolism, and transport along the intestinal tract. J. Agric. Food Chem. 2017, 65, 5480–5493. [Google Scholar] [CrossRef]
  99. Tomás-Barberán, F.A.; Seeram, N.P.; Espín, J.C. Bioavailability of pomegranate polyphenols. In Pomegranates: Ancient Roots to Modern Medicine; CRC Press: Boca Raton, FL, USA, 2006; Volume 3, pp. 45–60. [Google Scholar]
  100. García-Villalba, R.; Beltrán, D.; Espín, J.C.; Selma, M.V.; Tomás-Barberán, F.A. Time course production of urolithins from ellagic acid by human gut microbiota. J. Agric. Food Chem. 2013, 61, 8797–8806. [Google Scholar] [CrossRef]
  101. Heber, D. Herbal Medicine: Biomolecular and Clinical Aspects, 2nd ed.; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2011. [Google Scholar]
  102. Espín, J.C.; González-Barrio, R.; Cerdá, B.; López-Bote, C.; Rey, A.I.; Tomás-Barberán, F.A. Iberian pig as a model to clarify obscure points in the bioavailability and metabolism of ellagitannins in humans. J. Agric. Food Chem. 2007, 55, 10476–10485. [Google Scholar] [CrossRef]
  103. Gonzalez-Barrio, R.; Truchado, P.; Ito, H.; Espin, J.C.; Tomas-Barberan, F.A. UV and MS identification of urolithins and nasutins, the bioavailable metabolites of ellagitannins and ellagic acid in different mammals. J. Agric. Food Chem. 2011, 59, 1152–1162. [Google Scholar] [CrossRef]
  104. Espín, J.C.; Larrosa, M.; García-Conesa, M.T.; Tomás-Barberán, F. Biological significance of urolithins, the gut microbial ellagic acid-derived metabolites: The evidence so far. Evid. Based Complement. Altern. Med. 2013, 2013, 270418. [Google Scholar] [CrossRef]
  105. Nuñez-Sánchez, M.A.; García-Villalba, R.; Monedero-Saiz, T.; García-Talavera, N.V.; Gómez-Sánchez, M.B.; Sánchez-Álvarez, C.; García-Albert, A.M.; Rodríguez-Gil, F.J.; Ruiz-Marín, M.; Pastor-Quirante, F.A. Targeted metabolic profiling of pomegranate polyphenols and urolithins in plasma, urine and colon tissues from colorectal cancer patients. Mol. Nutr. Food Res. 2014, 58, 1199–1211. [Google Scholar] [CrossRef] [PubMed]
  106. Chang, W.-C.; Yu, Y.-M.; Chiang, S.-Y.; Tseng, C.-Y. Ellagic acid suppresses oxidised low-density lipoprotein-induced aortic smooth muscle cell proliferation: Studies on the activation of extracellular signal-regulated kinase 1/2 and proliferating cell nuclear antigen expression. Br. J. Nutr. 2008, 99, 709–714. [Google Scholar] [CrossRef]
  107. Zhao, W.; Wang, L.; Haller, V.; Ritsch, A. A novel candidate for prevention and treatment of atherosclerosis: Urolithin B decreases lipid plaque deposition in apoE−/− mice and increases early stages of reverse cholesterol transport in ox-LDL treated macrophages cells. Mol. Nutr. Food Res. 2019, 63, 1800887. [Google Scholar] [CrossRef] [PubMed]
  108. Rosenblat, M.; Volkova, N.; Aviram, M. Addition of pomegranate juice to statin inhibits cholesterol accumulation in macrophages: Protective role for the phytosterol beta-sitosterol and for the polyphenolic antioxidant punicalagin. Harefuah 2013, 152, 513. [Google Scholar]
  109. Park, S.-H.; Kim, J.-L.; Lee, E.-S.; Han, S.-Y.; Gong, J.-H.; Kang, M.-K.; Kang, Y.-H. Dietary ellagic acid attenuates oxidized LDL uptake and stimulates cholesterol efflux in murine macrophages. J. Nutr. 2011, 141, 1931–1937. [Google Scholar] [CrossRef] [PubMed]
  110. Rosenblat, M.; Volkova, N.; Aviram, M. Pomegranate phytosterol (β-sitosterol) and polyphenolic antioxidant (punicalagin) addition to statin, significantly protected against macrophage foam cells formation. Atherosclerosis 2013, 226, 110–117. [Google Scholar] [CrossRef] [PubMed]
  111. Cui, G.-H.; Chen, W.-Q.; Shen, Z.-Y. Urolithin A shows anti-atherosclerotic activity via activation of class b scavenger receptor and activation of nef2 signaling pathway. Pharmacol. Rep. 2018, 70, 519–524. [Google Scholar] [CrossRef] [PubMed]
  112. Andreux, P.A.; Blanco-Bose, W.; Ryu, D.; Burdet, F.; Ibberson, M.; Aebischer, P.; Auwerx, J.; Singh, A.; Rinsch, C. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat. Metab. 2019, 1, 595. [Google Scholar] [CrossRef]
  113. Yu, L.-M.; Dong, X.; Xue, X.-D.; Zhang, J.; Li, Z.; Wu, H.-J.; Yang, Z.-L.; Yang, Y.; Wang, H.-S. Protection of the myocardium against ischemia/reperfusion injury by punicalagin through an SIRT1-NRF-2-HO-1-dependent mechanism. Chem. Biol. Interact. 2019, 306, 152–162. [Google Scholar] [CrossRef]
  114. Lee, W.-J.; Ou, H.-C.; Hsu, W.-C.; Chou, M.-M.; Tseng, J.-J.; Hsu, S.-L.; Tsai, K.-L.; Sheu, W.H.-H. Ellagic acid inhibits oxidized LDL-mediated LOX-1 expression, ROS generation, and inflammation in human endothelial cells. J. Vasc. Surg. 2010, 52, 1290–1300. [Google Scholar] [CrossRef]
  115. Gao, H.; Huang, X.; Tong, Y.; Jiang, X. Urolithin B improves cardiac function and reduces susceptibility to ventricular arrhythmias in rats after myocardial infarction. Eur. J. Pharmacol. 2020, 871, 172936. [Google Scholar] [CrossRef] [PubMed]
  116. Han, Q.A.; Yan, C.; Wang, L.; Li, G.; Xu, Y.; Xia, X. Urolithin A attenuates ox-LDL-induced endothelial dysfunction partly by modulating microRNA-27 and ERK/PPAR-γ pathway. Mol. Nutr. Food Res. 2016, 60, 1933–1943. [Google Scholar] [CrossRef] [PubMed]
  117. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef]
  118. Xu, J.; Cao, K.; Liu, X.; Zhao, L.; Feng, Z.; Liu, J. Punicalagin regulates signaling pathways in inflammation-associated chronic diseases. Antioxidants 2021, 11, 29. [Google Scholar] [CrossRef]
  119. De Nigris, F.; Williams-Ignarro, S.; Sica, V.; Lerman, L.O.; D’Armiento, F.P.; Byrns, R.E.; Casamassimi, A.; Carpentiero, D.; Schiano, C.; Sumi, D. Effects of a pomegranate fruit extract rich in punicalagin on oxidation-sensitive genes and eNOS activity at sites of perturbed shear stress and atherogenesis. Cardiovasc. Res. 2007, 73, 414–423. [Google Scholar] [CrossRef]
  120. Rozentsvit, A.; Vinokur, K.; Samuel, S.; Li, Y.; Gerdes, A.M.; Carrillo-Sepulveda, M.A. Ellagic acid reduces high glucose-induced vascular oxidative stress through ERK1/2/NOX4 signaling pathway. Cell. Physiol. Biochem. 2017, 44, 1174–1187. [Google Scholar] [CrossRef] [PubMed]
  121. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef]
  122. Kojadinovic, M.; Arsic, A.; Petovic-Oggiano, G.; Gavrovic-Jankulovic, M.; Glibetic, M.; Popovic, M. Effect of urolithins on oxidative stress of colorectal adenocarcinomacells-Caco-2. Int. J. Food Sci. Nutr. 2017, 68, 952–959. [Google Scholar] [CrossRef]
  123. Ou, H.-C.; Lee, W.-J.; Lee, S.-D.; Huang, C.-Y.; Chiu, T.-H.; Tsai, K.-L.; Hsu, W.-C.; Sheu, W.H.-H. Ellagic acid protects endothelial cells from oxidized low-density lipoprotein-induced apoptosis by modulating the PI3K/Akt/eNOS pathway. Toxicol. Appl. Pharmacol. 2010, 248, 134–143. [Google Scholar] [CrossRef]
  124. Lee, G.; Park, J.-S.; Lee, E.-J.; Ahn, J.-H.; Kim, H.-S. Anti-inflammatory and antioxidant mechanisms of urolithin B in activated microglia. Phytomedicine 2019, 55, 50–57. [Google Scholar] [CrossRef]
  125. Silva-Islas, C.A.; Maldonado, P.D. Canonical and non-canonical mechanisms of Nrf2 activation. Pharmacol. Res. 2018, 134, 92–99. [Google Scholar] [CrossRef]
  126. Zheng, D.; Liu, Z.; Zhou, Y.; Hou, N.; Yan, W.; Qin, Y.; Ye, Q.; Cheng, X.; Xiao, Q.; Bao, Y. Urolithin B, a gut microbiota metabolite, protects against myocardial ischemia/reperfusion injury via p62/Keap1/Nrf2 signaling pathway. Pharmacol. Res. 2020, 153, 104655. [Google Scholar] [CrossRef]
  127. Lee, H.J.; Jung, Y.H.; Choi, G.E.; Kim, J.S.; Chae, C.W.; Lim, J.R.; Kim, S.Y.; Yoon, J.H.; Cho, J.H.; Lee, S.-J. Urolithin A suppresses high glucose-induced neuronal amyloidogenesis by modulating TGM2-dependent ER-mitochondria contacts and calcium homeostasis. Cell Death Differ. 2021, 28, 184–202. [Google Scholar] [CrossRef]
  128. Kiokias, S.; Proestos, C.; Oreopoulou, V. Effect of natural food antioxidants against LDL and DNA oxidative changes. Antioxidants 2018, 7, 133. [Google Scholar] [CrossRef]
  129. Atrahimovich, D.; Khatib, S.; Sela, S.; Vaya, J.; Samson, A.O. Punicalagin induces serum low-density lipoprotein influx to macrophages. Oxid. Med. Cell. Longev. 2016, 2016, 7124251. [Google Scholar] [CrossRef] [PubMed]
  130. Rosenblat, M.; Volkova, N.; Borochov-Neori, H.; Judeinstein, S.; Aviram, M. Anti-atherogenic properties of date vs. pomegranate polyphenols: The benefits of the combination. Food Funct. 2015, 6, 1496–1509. [Google Scholar] [CrossRef] [PubMed]
  131. Kang, I.; Kim, Y.; Tomás-Barberán, F.A.; Espín, J.C.; Chung, S. Urolithin A, C, and D, but not iso-urolithin A and urolithin B, attenuate triglyceride accumulation in human cultures of adipocytes and hepatocytes. Mol. Nutr. Food Res. 2016, 60, 1129–1138. [Google Scholar] [CrossRef] [PubMed]
  132. Cisneros-Zevallos, L.; Bang, W.Y.; Delgadillo-Puga, C. Ellagic acid and urolithins A and B differentially regulate fat accumulation and inflammation in 3T3-L1 adipocytes while not affecting adipogenesis and insulin sensitivity. Int. J. Mol. Sci. 2020, 21, 2086. [Google Scholar] [CrossRef]
  133. Altamimi, J.Z.; Alshammari, G.M.; AlFaris, N.A.; Alagal, R.I.; Aljabryn, D.H.; Albekairi, N.A.; Alkhateeb, M.A.; Yahya, M.A. Ellagic acid protects against non-alcoholic fatty liver disease in streptozotocin-diabetic rats by activating AMPK. Pharm. Biol. 2022, 60, 25–37. [Google Scholar] [CrossRef]
  134. Liu, R.; Li, J.; Cheng, Y.; Huo, T.; Xue, J.; Liu, Y.; Liu, J.; Chen, X. Effects of ellagic acid-rich extract of pomegranates peel on regulation of cholesterol metabolism and its molecular mechanism in hamsters. Food Funct. 2015, 6, 780–787. [Google Scholar] [CrossRef]
  135. Jean-Gilles, D.; Li, L.; Vaidyanathan, V.G.; King, R.; Cho, B.; Worthen, D.R.; Chichester Iii, C.O.; Seeram, N.P. Inhibitory effects of polyphenol punicalagin on type-II collagen degradation in vitro and inflammation in vivo. Chem-Biol. Interact. 2013, 205, 90–99. [Google Scholar] [CrossRef]
  136. Yaidikar, L.; Thakur, S. Punicalagin attenuated cerebral ischemia–reperfusion insult via inhibition of proinflammatory cytokines, up-regulation of Bcl-2, down-regulation of Bax, and caspase-3. Mol. Cell. Biochem. 2015, 402, 141–148. [Google Scholar] [CrossRef] [PubMed]
  137. Atrahimovich, D.; Samson, A.O.; Khattib, A.; Vaya, J.; Khatib, S. Punicalagin decreases serum glucose levels and increases PON1 activity and HDL anti-inflammatory values in Balb/c mice fed a high-fat diet. Oxid. Med. Cell. Longev. 2018, 2018, 2673076. [Google Scholar] [CrossRef] [PubMed]
  138. Komatsu, W.; Kishi, H.; Yagasaki, K.; Ohhira, S. Urolithin A attenuates pro-inflammatory mediator production by suppressing PI3-K/Akt/NF-κB and JNK/AP-1 signaling pathways in lipopolysaccharide-stimulated RAW264 macrophages: Possible involvement of NADPH oxidase-derived reactive oxygen species. Eur. J. Pharmacol. 2018, 833, 411–424. [Google Scholar] [CrossRef]
  139. Almowallad, S.; Huwait, E.; Al-Massabi, R.; Saddeek, S.; Gauthaman, K.; Prola, A. Punicalagin regulates key processes associated with atherosclerosis in THP-1 cellular model. Pharmaceuticals 2020, 13, 372. [Google Scholar] [CrossRef]
  140. Nguyen-Ngo, C.; Willcox, J.C.; Lappas, M. Anti-inflammatory effects of phenolic acids punicalagin and curcumin in human placenta and adipose tissue. Placenta 2020, 100, 1–12. [Google Scholar] [CrossRef]
  141. Yu, Y.-M.; Wang, Z.-H.; Liu, C.-H.; Chen, C.-S. Ellagic acid inhibits IL-1β-induced cell adhesion molecule expression in human umbilical vein endothelial cells. Br. J. Nutr. 2007, 97, 692–698. [Google Scholar] [CrossRef] [PubMed]
  142. Kannan, M.M.; Quine, S.D. Pharmacodynamics of ellagic acid on cardiac troponin-T, lyosomal enzymes and membrane bound ATPases: Mechanistic clues from biochemical, cytokine and in vitro studies. Chem. Biol. Interact. 2011, 193, 154–161. [Google Scholar] [CrossRef]
  143. Yoshimura, Y.; Nishii, S.; Zaima, N.; Moriyama, T.; Kawamura, Y. Ellagic acid improves hepatic steatosis and serum lipid composition through reduction of serum resistin levels and transcriptional activation of hepatic ppara in obese, diabetic KK-Ay mice. Biochem. Biophys. Res. Commun. 2013, 434, 486–491. [Google Scholar] [CrossRef]
  144. Zhang, Y.; Tan, X.; Cao, Y.; An, X.; Chen, J.; Yang, L. Punicalagin protects against diabetic liver injury by upregulating mitophagy and antioxidant enzyme activities. Nutrients 2022, 14, 2782. [Google Scholar] [CrossRef]
  145. Wang, Y.; Qiu, Z.; Zhou, B.; Liu, C.; Ruan, J.; Yan, Q.; Liao, J.; Zhu, F. In vitro antiproliferative and antioxidant effects of urolithin A, the colonic metabolite of ellagic acid, on hepatocellular carcinomas HepG2 cells. Toxicol. Vitr. 2015, 29, 1107–1115. [Google Scholar] [CrossRef] [PubMed]
  146. Lin, Z.; Lin, C.; Fu, C.; Lu, H.; Jin, H.; Chen, Q.; Pan, J. The protective effect of Ellagic acid (EA) in osteoarthritis: An in vitro and in vivo study. Biomed. Pharmacother. 2020, 125, 109845. [Google Scholar] [CrossRef] [PubMed]
  147. Zhou, E.; Fu, Y.; Wei, Z.; Yang, Z. Inhibition of allergic airway inflammation through the blockage of NF-κB activation by ellagic acid in an ovalbumin-induced mouse asthma model. Food Funct. 2014, 5, 2106–2112. [Google Scholar] [CrossRef] [PubMed]
  148. García-Villalba, R.; Giménez-Bastida, J.A.; Cortés-Martín, A.; Ávila-Gálvez, M.; Tomás-Barberán, F.A.; Selma, M.V.; Espín, J.C.; González-Sarrías, A. Urolithins: A comprehensive update on their metabolism, bioactivity, and associated gut microbiota. Mol. Nutr. Food Res. 2022, 66, e2101019. [Google Scholar] [CrossRef]
  149. Corrêa, T.A.F.; Rogero, M.M.; Hassimotto, N.M.A.; Lajolo, F.M. The two-way polyphenols-microbiota interactions and their effects on obesity and related metabolic diseases. Front. Nutr. 2019, 6, 188. [Google Scholar] [CrossRef]
  150. Selma, M.V.; Beltrán, D.; García-Villalba, R.; Espín, J.C.; Tomás-Barberán, F.A. Description of urolithin production capacity from ellagic acid of two human intestinal Gordonibacter species. Food Funct. 2014, 5, 1779–1784. [Google Scholar] [CrossRef]
  151. Cao, Y.; Ren, G.; Zhang, Y.; Qin, H.; An, X.; Long, Y.; Chen, J.; Yang, L. A new way for punicalagin to alleviate insulin resistance: Regulating gut microbiota and autophagy. Food Nutr. Res. 2021, 65. [Google Scholar] [CrossRef]
  152. Abdulrahman, A.O.; Alzubaidi, M.Y.; Nadeem, M.S.; Khan, J.A.; Rather, I.A.; Khan, M.I. Effects of urolithins on obesity-associated gut dysbiosis in rats fed on a high-fat diet. Int. J. Food Sci. Nutr. 2021, 72, 923–934. [Google Scholar] [CrossRef]
  153. Bialonska, D.; Ramnani, P.; Kasimsetty, S.G.; Muntha, K.R.; Gibson, G.R.; Ferreira, D. The influence of pomegranate by-product and punicalagins on selected groups of human intestinal microbiota. Int. J. Food Microbiol. 2010, 140, 175–182. [Google Scholar] [CrossRef]
  154. Viladomiu, M.; Hontecillas, R.; Lu, P.; Bassaganya-Riera, J. Preventive and prophylactic mechanisms of action of pomegranate bioactive constituents. Evid. Based Complement. Alternat. Med. 2013, 2013, 789764. [Google Scholar] [CrossRef]
  155. Aguilar, E.C.; Santos, L.C.; Leonel, A.J.; de Oliveira, J.S.; Santos, E.A.; Navia-Pelaez, J.M.; da Silva, J.F.; Mendes, B.P.; Capettini, L.S.; Teixeira, L.G.; et al. Oral butyrate reduces oxidative stress in atherosclerotic lesion sites by a mechanism involving NADPH oxidase down-regulation in endothelial cells. J. Nutr. Biochem. 2016, 34, 99–105. [Google Scholar] [CrossRef] [PubMed]
  156. Aguilar, E.C.; Leonel, A.J.; Teixeira, L.G.; Silva, A.R.; Silva, J.F.; Pelaez, J.M.; Capettini, L.S.; Lemos, V.S.; Santos, R.A.; Alvarez-Leite, J.I. Butyrate impairs atherogenesis by reducing plaque inflammation and vulnerability and decreasing NFκB activation. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 606–613. [Google Scholar] [CrossRef]
  157. O’Morain, V.L.; Chan, Y.H.; Williams, J.O.; Alotibi, R.; Alahmadi, A.; Rodrigues, N.P.; Plummer, S.F.; Hughes, T.R.; Michael, D.R.; Ramji, D.P. The Lab4P consortium of probiotics attenuates atherosclerosis in LDL receptor deficient mice fed a high fat diet and causes plaque stabilization by inhibiting inflammation and several pro-atherogenic processes. Mol. Nutr. Food Res. 2021, 65, e2100214. [Google Scholar] [CrossRef] [PubMed]
  158. Al-Ahmadi, W.; Webberley, T.S.; Joseph, A.; Harris, F.; Chan, Y.H.; Alotibi, R.; Williams, J.O.; Alahmadi, A.; Decker, T.; Hughes, T.R.; et al. Pro-atherogenic actions of signal transducer and activator of transcription 1 serine 727 phosphorylation in LDL receptor deficient mice via modulation of plaque inflammation. FASEB J. 2021, 35, e21892. [Google Scholar] [CrossRef] [PubMed]
  159. Ramji, D.P.; Chan, Y.H.; Alahmadi, A.; Alotibi, R.; Alshehri, N. Survey of approaches for investigation of atherosclerosis in vivo. Methods Mol. Biol. 2022, 2419, 57–72. [Google Scholar]
  160. Ramji, D.P.; Ismail, A.; Chen, J.; Alradi, F.; Al Alawi, S. Survey of in vitro model systems for investigation of key cellular processes associated with atherosclerosis. Methods Mol. Biol. 2022, 2419, 39–56. [Google Scholar]
  161. Moss, J.W.E.; Williams, J.O.; Al-Ahmadi, W.; O’Morain, V.; Chan, Y.H.; Hughes, T.R.; Menendez-Gonzalez, J.B.; Almotiri, A.; Plummer, S.F.; Rodrigues, N.P.; et al. Protective effects of a unique combination of nutritionally active ingredients on risk factors and gene expression associated with atherosclerosis in C57BL/6J mice fed a high fat diet. Food Funct. 2021, 12, 3657–3671. [Google Scholar] [CrossRef]
Figure 1. Transformation of punicalagin to urolithins. Food-derived ellagitannins, such as punicalagin in pomegranates, are first hydrolyzed in the stomach to produce ellagic acid. This then undergoes a series of transformation by the gut microbiota to produce the different urolithins with urolithin-A and -B being two key final products. Figure created using Biorender.com, accessed on 28 March 2023.
Figure 1. Transformation of punicalagin to urolithins. Food-derived ellagitannins, such as punicalagin in pomegranates, are first hydrolyzed in the stomach to produce ellagic acid. This then undergoes a series of transformation by the gut microbiota to produce the different urolithins with urolithin-A and -B being two key final products. Figure created using Biorender.com, accessed on 28 March 2023.
Ijms 24 08476 g001
Figure 2. The biological effects of PC and its metabolites on atherosclerosis. ↑, increased/improved; ↓, decreased; EA, ellagic acid; oxLDL, oxidized low density lipoprotein; PC, punicalagin; UroA, urolithin A; UroB, urolithin B; UroC, urolithin C; UroD, urolithin D. Figure created using Biorender.com, accessed on 28 March 2023.
Figure 2. The biological effects of PC and its metabolites on atherosclerosis. ↑, increased/improved; ↓, decreased; EA, ellagic acid; oxLDL, oxidized low density lipoprotein; PC, punicalagin; UroA, urolithin A; UroB, urolithin B; UroC, urolithin C; UroD, urolithin D. Figure created using Biorender.com, accessed on 28 March 2023.
Ijms 24 08476 g002
Figure 3. Effect of PC and its metabolites on various factors influencing atherogenesis and non-alcoholic fatty liver disease progression. Both atherosclerosis and NAFLD/NASH are caused, and exacerbated by, a number of similar factors, including diet, stress, insulin resistance and genetic disorders, combined with an increase in triacylglycerol, reactive oxygen species, and tumour necrosis factor-α levels. These can then cause the development of NAFLD/NASH in the liver, which further contributes to the production of pro-inflammatory cytokines, such as interleukin-1β and -6, or elevated triacylglycerol and reactive oxygen species, which can all contribute to the development of atherosclerosis. PC and its metabolites can decrease the impact of these factors and represent the prevention or therapeutic approach to NAFLD/NASH and atherosclerosis. ↑, increased; IL, interleukin; LDL, low-density lipoprotein; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; ROS, reactive oxygen species; TG, triacylglycerol; TNF, tumour necrosis factor. Created using BioRender.com, accessed on 28 March 2023.
Figure 3. Effect of PC and its metabolites on various factors influencing atherogenesis and non-alcoholic fatty liver disease progression. Both atherosclerosis and NAFLD/NASH are caused, and exacerbated by, a number of similar factors, including diet, stress, insulin resistance and genetic disorders, combined with an increase in triacylglycerol, reactive oxygen species, and tumour necrosis factor-α levels. These can then cause the development of NAFLD/NASH in the liver, which further contributes to the production of pro-inflammatory cytokines, such as interleukin-1β and -6, or elevated triacylglycerol and reactive oxygen species, which can all contribute to the development of atherosclerosis. PC and its metabolites can decrease the impact of these factors and represent the prevention or therapeutic approach to NAFLD/NASH and atherosclerosis. ↑, increased; IL, interleukin; LDL, low-density lipoprotein; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis; ROS, reactive oxygen species; TG, triacylglycerol; TNF, tumour necrosis factor. Created using BioRender.com, accessed on 28 March 2023.
Ijms 24 08476 g003
Table 1. The main biological effects of PC and its metabolites in vitro and in vivo.
Table 1. The main biological effects of PC and its metabolites in vitro and in vivo.
MarkersAgentDurationModelReference
oxLDL levels or oxLDL-mediated responses ↓PC (195 mg/day)20 weeksHuman: healthy individuals, 45–65 years[88]
EA (50 µM) on oxLDL-induced proliferation24 hRat thoracic smooth muscle cells[106]
UroB (0.1–10 μM)24 hHuman THP-1 macrophages[107]
Cholesterol Efflux ↑PC (10–26 µM)20 hMurine J774A.1 macrophages[108]
EA (1–5 µM)24 hMurine J774A.1 macrophages[109]
UroB (0.1–10 µM)24 hMurine J774A.1 macrophages[107]
Cholesterol Biosynthesis ↓ PC (15 or 30 µM)20 hMurine J774A.1 macrophages[110]
Plasma Lipids ↓UroA (3 mg/kg/day)12 weeks Adult Wister rats [111]
Fatty Acid Oxidation ↓UroA (250–2000 mg/day)28 daysHuman: healthy individuals (61–85 years)[112]
Inflammatory cytokines/markers ↓ PC (40 mg/kg/day)3 daysSprague-Dawley rats[113]
EA (5–20 µM)2 h and then 150 μg/mL oxLDL for 24 hHuman umbilical cord endothelial cells [114]
UroA (18 µM)4–12 hTNF-α-activated human aortic endothelial cells[94]
UroB (2.5–5 mg/kg/day)2 weeksAdult male Sprague Dawley rats[115]
Autophagy/Mitophagy and mitochondrial health ↑UroA (250–2000 mg/day)4 weeksHuman: Healthy individuals (61–85 years) [112]
Endothelial function ↑PC (195 mg/day)20 weeks Human: Healthy individuals aged 45–65 years[88]
UroA (0.5–5 µM)24 hoxLDL-treated human aortic endothelial cells[116]
Plaque lipid deposition ↓UroB (10 mg/kg/day)14 daysMale Apo-E−/− mice[107]
↑, increased/improved; ↓, decreased; ApoE−/−, apolipoprotein E deficient mice; EA, ellagic Acid; oxLDL, oxidized low density lipoprotein; PC, Punicalagin; TNF-α, tumour necrosis factor-α; UroA, urolithin A; UroB, urolithin B.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alalawi, S.; Albalawi, F.; Ramji, D.P. The Role of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease. Int. J. Mol. Sci. 2023, 24, 8476. https://doi.org/10.3390/ijms24108476

AMA Style

Alalawi S, Albalawi F, Ramji DP. The Role of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease. International Journal of Molecular Sciences. 2023; 24(10):8476. https://doi.org/10.3390/ijms24108476

Chicago/Turabian Style

Alalawi, Sulaiman, Faizah Albalawi, and Dipak P. Ramji. 2023. "The Role of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease" International Journal of Molecular Sciences 24, no. 10: 8476. https://doi.org/10.3390/ijms24108476

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