Next Article in Journal
Potential of Natural Phenolic Compounds against Doxorubicin-Induced Chemobrain: Biological and Molecular Mechanisms Involved
Previous Article in Journal
Hormetic Nutrition and Redox Regulation in Gut–Brain Axis Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unlocking the Therapeutic Potential of Ellagic Acid for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis

by
Tharani Senavirathna
1,
Armaghan Shafaei
2,
Ricky Lareu
3,* and
Lois Balmer
1,*
1
Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA 6027, Australia
2
Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Perth, WA 6027, Australia
3
Curtin Medical School and Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, WA 6845, Australia
*
Authors to whom correspondence should be addressed.
Antioxidants 2024, 13(4), 485; https://doi.org/10.3390/antiox13040485
Submission received: 5 March 2024 / Revised: 8 April 2024 / Accepted: 17 April 2024 / Published: 18 April 2024
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)

Abstract

:
Obesity is in epidemic proportions in many parts of the world, contributing to increasing rates of non-alcoholic fatty liver disease (NAFLD). NAFLD represents a range of conditions from the initial stage of fatty liver to non-alcoholic steatohepatitis (NASH), which can progress to severe fibrosis, through to hepatocellular carcinoma. There currently exists no treatment for the long-term management of NAFLD/NASH, however, dietary interventions have been investigated for the treatment of NASH, including several polyphenolic compounds. Ellagic acid is one such polyphenolic compound. Nutraceutical food abundant in ellagic acid undergoes initial hydrolysis to free ellagic acid within the stomach and small intestine. The proposed mechanism of action of ellagic acid extends beyond its initial therapeutic potential, as it is further broken down by the gut microbiome into urolithin. Both ellagic acid and urolithin have been found to alleviate oxidative stress, inflammation, and fibrosis, which are associated with NAFLD/NASH. While progress has been made in understanding the pharmacological and biological activity of ellagic acid and its involvement in NAFLD/NASH, it has yet to be fully elucidated. Thus, the aim of this review is to summarise the currently available literature elucidating the therapeutic potential of ellagic acid and its microbial-derived metabolite urolithin in NAFLD/NASH.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is the physiological manifestation of obesity in the liver. This fast-growing epidemic is the most prevalent form of chronic liver disease globally. The prevalence of NAFLD has increased from 25.24% in 2015 to 29.38% in 2021 [1,2,3]. This condition now accounts for 45.8% of all cases of chronic-liver-disease-related deaths worldwide [1,3,4]. NAFLD can advance to non-alcoholic steatohepatitis (NASH), and approximately 20% of the affected population is likely to develop NASH [1,5]. Hepatic steatosis, alongside hepatocellular injury and inflammation, define NASH as these are the significant contributors to the onset of cirrhosis and hepatocellular carcinoma [1,6]. Although the specific pathogenesis of NASH remains uncertain, associated risk factors such as excess caloric intake, sedentary lifestyle, insulin resistance, liver lipogenesis, and gut microbiota dysbiosis are well established [7,8,9,10]. Despite years of extensive global research, there is currently no approved drug for the treatment of NASH [5,10,11,12]. Oxidative stress significantly contributes to the progression from NAFLD to NASH evidenced by an increase in oxidative stress and impaired antioxidant defence mechanisms throughout disease progression [7,13]. Several studies have been undertaken to investigate the efficacy of antioxidants in mitigating this phenomenon [10,14,15,16]. Ellagic acid is widely recognized for its antioxidant properties, but it also exhibits anti-inflammatory, antifibrotic, and anticancer properties [17,18,19,20]. In recent decades, this common non-flavonoid polyphenolic compound caught the attention for its hepatoprotective properties and as a therapeutic agent for treating NAFLD/NASH [14,21,22]. This review focuses on the effectiveness of ellagic acid in treating NAFLD/NASH by summarising relevant literature concerning its potential therapeutic mechanism on the liver.

2. Current Understanding of NAFLD/NASH

NAFLD represents a range of conditions from simple fatty liver (non-alcoholic fatty liver, NAFL) to NASH, which can progress to more severe fibrosis, cirrhosis, and potentially liver cancer [9,23]. NAFLD is characterised by over-accumulation of triglycerides in hepatocytes and is actively involved in all aspects of the Metabolic Syndrome, including obesity, type 2 diabetes mellitus, arterial hypertension, and hyperlipidaemia [24,25,26]. Progression to NASH is characterised by active liver tissue damage through increased inflammation and hepatocyte ballooning, as measured by the NAFLD Activity Score (NAS) [6].
Research advocates have campaigned for the renaming of NAFLD to metabolic dysfunction associated with fatty liver disease (MAFLD), arguing that NAFLD is viewed as an exclusionary term, defined only in the absence of conditions such as viral hepatitis B and C, autoimmune disorders, or excessive alcohol consumption [27,28]. In contrast to NAFLD, MAFLD is diagnosed on the onset of hepatic steatosis alongside the presence of one of the following: obesity/overweight, diabetes mellitus, or indicators of metabolic dysregulation [29,30]. Globally, MAFLD affects 38.8% of adults, carrying the potential to progress to cirrhosis and instigate significant extrahepatic conditions such as cardiovascular disease and chronic kidney disease [31,32]. However, the diagnostic criteria and the concept are novel and yet to be further tested and validated.

Manifestation of NAFLD/NASH

The current understanding of NAFLD/NASH development and progression has shifted from the traditional two-hit hypothesis to a multiple, parallel-hit hypothesis, where pathogenetic influences act synergistically [8]. According to the multiple-hit hypothesis, hepatocellular damage not only originates from insulin resistance but also from dysbiosis of the gut microbiota, overnutrition, secretion of hormones from adipose tissue, and genetic and epigenetic factors [7,8]. Insulin resistance plays a pivotal role in altering lipid metabolism in the body, where it stimulates hepatic de novo lipogenesis and adipose tissue lipolysis, causing an escalated influx of fatty acids to the liver [33,34,35,36]. This results in elevated levels of inflammatory cytokine and adipokine secretion due to adipose tissue dysfunction [34,37]. Adipokines such as adiponectin and leptin induce insulin resistance, stimulate lipogenesis, and trigger inflammation, resulting in the accumulation of triglycerides within hepatocytes and their damage [38,39,40]. Cytokines play a crucial role in promoting inflammation in the liver. Pro-inflammatory cytokines such as tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 β (IL-1β) activate and enhance liver inflammation, leading to hepatic inflammation and hepatocyte damage [41,42,43]. Intrahepatic accumulation of fatty acids induces the production of lipotoxic lipids, which then increase endoplasmic reticulum stress and mitochondrial dysfunction. More importantly, in the liver, this results in an increase in oxidative stress and inflammation, causing the progression of NASH from steatosis [11,44,45]. Dysbiosis of the gut microbiome has also been found to activate the innate immune system, causing a cascade of inflammatory cytokines to be released from immune cells [46,47,48,49,50]. Subsequently, the gut microbiome has been identified as a key player in the progression of NAFLD to NASH; however, further research is needed to fully understand the role of the gut microbiome. One such mechanism of action is dysbiosis of the gut microbiome, which alters bile acid production and composition [51,52,53]. Alterations in bile acids can damage hepatocytes by promoting cellular injury, apoptosis, mitochondrial dysfunction, membrane disruption, and oxidative stress leading to liver damage [54,55,56]. These interactions further add to the hepatocellular damage, propelling the progression of NAFLD to NASH [7,8,9,57].
An increase in oxidative stress has been identified to be a major accelerant in the progression of NAFLD to NASH [58,59]. Malfunction of cellular organelles, such as the mitochondria and endoplasmic reticulum drives the increase in oxidative stress seen with liver damage [44,60]. During the progression of liver disease, uncontrolled accumulation of free fatty acids and lipotoxic lipids leads to an increase in reactive oxygen species and the exhaustion of the antioxidant defence system, which includes both enzymes and non-enzymatic antioxidants in the liver, which in turn may cause changes in hepatocyte structure and function [61,62]. Collectively, these processes result in chronic inflammation followed by hepatocyte cell death and hepatic stellate cell (HSC) activation, which then leads to fibrosis [45,59,60,63,64]. At the cellular level, the development of NASH is characterised by interactions between resident liver cells and immune-recruited cells, including liver progenitor cells (LPC), HSCs, and macrophages [26,65,66,67,68].

3. Current Treatments

There are no specific medications approved by the European Medicines Agency (EMA) or the Food and Drug Administration (FDA) for the treatment of NAFLD/NASH [10,12,69,70]. Lifestyle changes that include healthier eating and regular exercise are an effective treatment for NASH [71,72], along with weight loss and bariatric surgery [10,65,71,73,74,75]. Increased intake of carbohydrates, trans- and saturated fats, animal proteins (red meat), and processed food coupled with low intake of fibrous food are linked with NAFLD/NASH progression [76,77]. Portion control and calorie restriction are important, as excessive calorie consumption can lead to weight gain and exacerbate liver fat accumulation [78,79]. Recommendations suggest focusing on whole grains and low-glycemic index foods for carbohydrate intake and prioritising monounsaturated and polyunsaturated fats over other types for dietary fat intake [77,79,80,81,82]. Additionally, integrating vegetable protein sources, prebiotic fibers, and probiotic-enriched foods into the diet not only aids in reducing calorie intake but also fosters healthy gut microbiota [76,83,84]. Moreover, incorporating antioxidant-rich foods like berries, nuts, and leafy greens can mitigate oxidative stress and inflammation, crucial factors contributing to liver damage in NAFLD/NASH [78,84,85]. However, unlike communicable diseases, these treatments experience high dropout rates, a common phenomenon observed with other therapies that require individual behavioural changes. The dropout rate for lifestyle-based treatments, including diet change and greater physical activity has been reported to be between 10–80% [86]. These findings illustrate the necessity for effective treatments utilising pharmacological agents, both as a complement to lifestyle modifications and as a standalone approach for managing the significant proportion of patients that are unable to adopt “healthier” lifestyles due to mental and physical impairments, as well as busy lifestyles.
Several therapeutic approaches have been proposed for the treatment of NASH, however, none have yet been approved [10,12,87,88]. Recent studies indicate that antidiabetic drugs and glucagon-like peptide 1 (GLP-1) receptor agonists represent promising treatments for NASH [89,90]. Two types of agonists, Liraglutide and Semaglutide, have been shown to be effective in reducing insulin resistance, liver lipotoxicity, and hyperglycemia in patients with NASH [89,91,92]. However, a lack of oral administration and an increased risk of pancreatitis are two known side effects of these drugs [12].
Semaglutide, an FDA-approved treatment for type 2 diabetes, is recognised for its greater metabolic effects compared to Liraglutide. In a randomized, placebo-controlled, phase 2 clinical trial, researchers investigated Semaglutide impact on the histologic resolution of NASH in patients diagnosed with biopsy-confirmed NASH and fibrosis [93]. The findings revealed that a significantly higher percentage of patients experienced NASH resolution with Semaglutide compared to the placebo (59% with the 0.4-mg dose vs. 17% with the placebo), but there was no significant difference observed in improving fibrosis [93]. The study is currently progressing to the next phase, with an ongoing phase 3 trial (NCT04822181). Several clinical trials have shown that peroxisome proliferator-activated receptor (PPAR)-γ agonists, such as pioglitazone, significantly reduce steatosis, inflammation, and fibrosis, whilst improving levels of plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) [94,95,96,97]. However, side effects such as weight gain, bone loss in women, and possible bladder cancer impede the use of these drugs in the treatment of NASH [95]. Lanifibranor acts as an oral pan-PPAR agonist. A successfully conducted 2b double-blind, randomised, placebo-controlled clinical trial on Lanifibranor [98] identified that patients administered a once-daily dose of 1200 mg demonstrated a significantly higher reduction percentage (55% vs. 33% for placebo) in the SAF-A score (representing the activity component of Steatosis, Activity, Fibrosis) with no exacerbation of fibrosis compared to the placebo group [98]. The study has progressed to the phase 3 trial (NCT03008070).
Recent findings suggest that vitamin E treatment may be more suitable for paediatric patients with NASH compared to adults [99], targeting oxidative stress and inflammation [95,100]. However, long-term use of vitamin E has been associated with increased risks of prostate cancer and haemorrhagic stroke [101,102,103]. In a randomised, controlled trial spanning two years, non-diabetic patients with NASH were administered either vitamin E (800 IU, natural form, once daily), pioglitazone (30 mg once daily), or a placebo, revealing that vitamin E improved NASH in 43% of patients, while pioglitazone showed no significant effect compared to placebo. Currently, both pioglitazone and vitamin E are used on a case-by-case basis, as a comprehensive understanding of the risk factors is yet to be thoroughly understood [2,95,104].
Resmetirom has recently emerged as a promising candidate for the treatment of NAFLD/NASH. As an agonist for the thyroid hormone receptor-β, Resmetirom aims to address key metabolic pathways compromised during the progression of NAFLD/NASH, including lipid metabolism, fibrosis, and inflammation [105]. Results from a randomised, double-blinded, placebo-controlled phase 2 clinical trial demonstrated significant efficacy of resmetirom compared to placebo over 12 and 36 weeks. Adult patients exhibited a significantly greater relative reduction in hepatic fat (32·9% with resmetirom vs 10·4% with placebo) as assessed by liver biopsy [106]. The most recent randomised, double-blinded, placebo-controlled phase 3 clinical study (NCT03900429) corroborates these findings, indicating that resmetirom is safe and well-tolerated in NASH patients over 52 weeks [107].

4. Ellagic Acid and Dietary Sources

Ellagic acid (EA) is a polyphenolic, non-flavonoid compound naturally found in a variety of fruits, such as pomegranates, raspberries, strawberries, and grapes, and nuts, such as pistachios, pecans, walnuts, and acorns [17,21,108,109]. Ellagic acid is a dilactone with the chemical name 2,3,7,8-Tetrahydroxy [1] benzopyrano [5,4,3-cde][1]benzopyran-5,10-dione (C14H6O8; MW: 302.194 g/mol; CAS number, 476-66-4) possessing both a hydrophilic moiety with four hydroxyl groups and two lactone groups together with a lipophilic moiety with two hydrocarbon rings [110]. This structure enables EA to accept electrons from several substrates, thereby participating in antioxidant redox reactions [110,111]. EA is commercially available as a nutraceutical product and known to attenuate chronic diseases such as metabolic syndrome, cardiovascular disease, hypertension, and neurodegenerative diseases [112,113,114,115].

4.1. Ellagic Acid Metabolites

Dietary EA is presented in free form or as a hydrolysable complex polymer called ellagitannins, which can be further metabolized to release free EA and gallic acid [18]. Free EA is primarily absorbed by the stomach and small intestine. The remainder is either absorbed by the large intestine [17,116,117] or metabolised by the resident gut microbiota to produce a microbial-derived metabolite known as urolithin [18,118,119]. In the liver, EA undergoes phase I metabolism, which involves oxidation and hydrolysis followed by phase II metabolism [18]. During phase II metabolism, ellagic acid undergoes a series of reactions, including glucuronidation, sulfation, and methylation. This results in the formation of ellagic acid glucuronides, methyl ellagic acid glucuronides, and dimethyl ellagic acid glucuronide, which are found in bile, confirming enterohepatic circulation [18,118,120,121].
Ellagitannins are resistant to acid hydrolysis and, as such, are not directly absorbed in the stomach [122,123]. Hydrolysation and absorption of ellagitannins occurs in the small intestine, under a slightly basic or natural pH, which enables further absorption of free EA. EA is then absorbed across the gut epithelium through passive diffusion [17,124]. Unabsorbed free EA and ellagitannins are further metabolized and absorbed in the later part of the gastrointestinal tract. Urolithins are produced by gut microbiota during the metabolism of EA in humans [125,126] and several animals including rats [117] and mice [127,128]. Urolithin is a dibenzopyran-6-one derivative, which is also considered a benzo coumarin or dibenz-œ-pyrone [129]. During the microbial metabolism of EA, urolithin D is first produced via lactone ring cleavage and decarboxylation. Then, urolithin D is dehydroxylated from urolithin C, urolithin A, isourolithin A, and urolithin B, respectively (Figure 1) [17,20,129]. There are three main human metabotypes of urolithin, namely, metabotypes 0 (no urolithin production), metabotypes A (urolithin A), and metabotypes B (urolithin B, isourolithin A, and urolithin A) [119]. However, a study by Garcia-Villalba and colleagues revealed a new urolithin metabolic branch named R [130]. EA metabolism and urolithin production vary according to host health, age, gut microbial composition, environmental conditions, and metabotypes [14,119,131,132]. The presence of specific bacterial genera such as Gordonibacter and Ellagibacter has been shown to be necessary for the conversion of EA into urolithins [118,119,133,134]. Two gut microbial species, Gordonibacte urolithinfaciens and Gordonibacte pamelaeae, can produce urolithin C, whereas Ellagibacter isourolithinfaciens produces isourolithin A [132,135,136].

4.2. Bioavailability of Ellagic Acid, Ellagitannins, and Urolithins

The low bioavailability of ingested EA and its metabolites makes it difficult to detect and fully understand its metabolism. The level of EA in serum is very low (~200 ng mL−1) [137] but urolithins and their derivatives are present in micromolar levels, ranging from 0.024 to 35 μM in human plasma [118,120]. One glass of pomegranate juice (237 mL) can yield up to 300 mg of ellagitannins or about 120 mg of EA [138]. Around 100 g of raspberries produces 300 mg of ellagitannins and one strawberry yield up to 70 mg of ellagitannins [139]. However, urolithins have a greater absorption value than EA, possessing a higher lipo-solubility than free EA and, therefore, are more readily absorbed [17,140,141]. A study that investigated the metabolites present in defatted walnut powder administered to male Sprague Dawley rats revealed the presence of urolithin M5, urolithin C, and urolithin D [142]. Urolithin M5 is the first urolithin formed by the opening of one of the two lactone rings of ellagic acid, followed by decarboxylation [118,130]. These compounds were identified as hydrolyzation and dehydroxylation products of EA following the administration of defatted walnut power extract at a dosage of 10 g/kg for two days [142]. Interestingly, the study also detected both methylated and demethylated metabolites of EA in urine and faeces samples collected from the rats [142]. Analysis of blood and urine samples across several clinical studies indicated that the maximum concentration of EA occurred within 1–2 h and 0–4 h following gastrointestinal administrating [21,137,138,143]. This rapid elimination of EA presents a significant challenge in comprehending its biological functions, biotransformation, and bioavailability in animal and human systems. Consequently, measuring EA and its metabolites allows researchers to identify potential targeted biomarkers and gain a deeper understanding of EA biological activity.

5. Mechanisms of Action

Ellagic acid and its microbial metabolite, the urolithins, have been reported to possess many beneficial pharmacological properties such as antioxidant, anti-inflammatory, antimutagenic, antidepressant, cardio-protectant, anticarcinogenic, and most of most relevance to this review, hepatoprotective activity (Figure 2) [18,22,109,144].

5.1. Oxidative Stress

EA is a recognised antioxidant with implications in modulating various molecular targets and pathways involved in numerous chronic diseases, including liver diseases [17,109,145]. With its four hydroxyl and two lactone functional groups, EA exhibits the capacity to scavenge a wide variety of free radical species, including reactive oxygen species (ROS) and reactive nitrogen species (NOS); thus, protecting against free radical-induced damage [145,146,147,148].
The presence of hydroxyl and peroxyl radicals enables the initiation and multiplication of lipid peroxidation, respectively. Studies have demonstrated the efficacy of EA in mitigating lipid peroxidation even at minimal concentrations, such as µM levels, which is primarily attributed to its potent free radical scavenging properties [149]. Interestingly, an earlier investigation proposed that EA was a superior free radical scavenger to vitamin E [150]. Recently, it has been reported that EA inhibited 71.2% of lipid peroxidation, surpassing the inhibition levels observed with well-established antioxidants, ascorbic acid and α-tocopherol (i.e., vitamin E), resulting in 64.5% and 59.7% inhibition, respectively [151]. According to Yu and colleagues [152], EA supplementation in rabbits fed an atherogenic diet led to significant improvements in their lipid profiles and a reduction in lipid peroxidation. This intervention also resulted in the suppression of 8-oxo-2′-deoxyguanosin (8-(OH)dG) levels, in addition to the expression of caspase-8, caspase-9, and Fas ligand in the aortic arch. These findings suggest that the reduction in lipid peroxidation contributes to the antioxidant effects of EA.
The presence of Ionic cations such as Zn2+, Ca2+, Fe2+, Cd2+, and Cu2+ can promote oxidation rates in a biological system. However, phenolic compounds such as EA can inhibit this process due to its chelating property, where, it binds to the cation and forms a complex that prevents oxidation [153,154]. Ahmed and colleagues [155] have previously described how EA (500 µmol/Kg BW(body weight)) has a chelating effect that suppresses nickel-induced oxidative stress in female Wistar rats. Furthermore, Kilic and co-authors [151] have reported that EA has a similar chelating effect as caffeic acid on ferrous ions. Moreover, EA and epigallocatechin gallate have a similar binding capability to iron, where this binding is enabled due to the presence of catechol groups in the former [156]. EA also can chelate copper and form stable complexes after deprotonation [157]. Collectively, these data demonstrate that EA is protected from oxidative stress by chelation.
The formation of 8-(OH)dG is a critical step in oxidative damage on DNA, and several studies have reported that EA drastically reduces 8-(OH)dG formed by oxidative DNA damage [158,159]. Previous in vitro studies have described this protective effect of EA via its ability to regulate intracellular mechanisms through direct interaction between double-strand DNA and EA [160,161]. An in vitro study by Spencer and colleagues [162] has revealed that EA has the ability to inhibit dopamine/Cu2+-induced oxidative DNA damage even at very low doses, as low as 1 µM. This suggests EA’s potential protective mechanism against oxidative stress through shielding DNA damage.
Nuclear factor erythroid 2–related factor (Nrf2) is a cellular antioxidant regulator that activates during cellular stress to induce genes related to the antioxidant defence system. Activation of these types of responses or signalling pathways plays a major role in protective mechanisms against oxidative stress [163,164]. In this regard, EA administration to high-fat diet (HFD)-fed apolipoprotein E-knockout mice (ApoE−/−), resulted in a reduction in oxidative stress and atherosclerosis via induction of the Nrf2 signalling pathway [165]. In vitro research conducted by Baek and colleagues [166] reported that EA upregulates the Nrf2 pathway in human dermal fibroblasts, this then plays a protective role against induced oxidative stress. Furthermore, another study by Gu and co-authors [167] has described the protective effect of EA in acute hepatic injury in mice, by inducing Nrf2 expression and heme oxygenase-1. These studies conclude that EA has protective properties against oxidative stress through upregulation of the Nrf2 pathway.

5.2. Inflammation

In addition to its well-known antioxidant properties, EA has been reported to possess anti-inflammatory properties [168]. As mentioned above, both oxidative stress and inflammation are closely associated [169,170]. Nuclear factor kappa B (Nf-kB) is a key transcription factor for proinflammatory responses, produced in all cell types and activated under several types of cell stress induced by obesity, oxidative stress, hyperglycaemia, hypertension, and bacterial infections [171,172,173]. A study by Ahad and colleagues [174] has reported EA supplementation attenuated dyslipidaemia and nephropathy in type 2 diabetic male Wistar rats. The authors described how EA ameliorated diabetic nephropathy by inhibiting the expression of the Nf-kB pathway. Nf-kB has also been reported to play a key role in the regulation of cyclooxygenase-2 (COX-2) expression, which is involved in the inflammatory process during tumour growth [175]. Administration of EA (100 mg/Kg BW) can modulate COX-2 mRNA, mainly through downregulation of ROS production, which in turn inhibits Nf-kB activation [176].
Different kinds of proinflammatory cytokines, such as macrophages and migration inhibitory factor (MIF), play a crucial role in facilitating an immune response [109]. It has been shown that MIF induces Nf-kB and chemotaxis during an inflammatory response [109,177,178,179]. EA (50 µM) downregulated the tautomerase activity of MIF and MIF-mediated proinflammatory responses in peripheral blood mononuclear cells [179]. EA also suppresses the expression of pro-inflammatory cytokines TNFα, IL-6, and chemokine C-C, in lipopolysaccharide (LPS)-stimulated macrophages and adipocytes, suggesting that EA may attenuate inflammation in adipose tissue [180]. Moreover, EA can significantly inhibit TNFα and IL-6 in LPS-stimulated RAW 264.7 cells, even with minimal µM concentrations (6.25 µM and 12.5 µM) [181]. Even though the exact mechanism of EA involvement in proinflammatory cytokine modulation is not clear, it has been proposed that this polyphenol acts through direct inhibition of the Nf-kB pathway.
Another mechanism of interest is the effect of EA on resistin, an adipocytokine which may be the missing link between obesity and type 2 diabetes [182,183]. Pomegranate fruit extract suppressed enhanced levels of serum resistin in mice [184]. The authors also showed that EA reduced resistin levels in 3T3-L1 cells in vitro. Another study by the same authors demonstrated that EA reduced serum resistin levels without changing mRNA expression in adipose tissue. Further to this, they demonstrated that EA significantly improved hepatic steatosis and serum lipid composition in KK-Ay mice, thus further contributing to EA in the suppression of resistin secretion in vivo [185].

6. Therapeutic Efficacy

Recent studies, both in vivo and in vitro, have shown that EA forms several natural metabolites that have potential therapeutic properties for chronic diseases such as liver disease [18,21,109]. Research has focused on this nutraceutical due to its pharmacokinetic properties, safety, and efficiency [109,186,187]. A study conducted to determine the role of muscadine grape extract vs muscadine grape wine in obesity has demonstrated that both supplementations reduced plasma triglycerides, free fatty acids, and cholesterol levels [188]. However, muscadine grape extract also demonstrated a higher lipid-lowering effect on both triglycerides content and adipose tissue mass and possesses a higher content of EA (18 mg EA/Kg BW) than the wine. The wine, which has similar polyphenolic content but lower EA content (1.1 mg EA/Kg BW), demonstrated lower metabolic improvements, suggesting EA might play a crucial role in improving the Metabolic Syndrome.
The involvement of gene regulation in fatty acid oxidation has been studied by Cao and colleagues [189], demonstrating that supplementation of punicalagin (150 mg EA/Kg BW), one of the main ellagitannins in pomegranate, attenuated HFD-induced obesity in rats through mechanisms involving AMP-activated protein kinase (AMPK) activation [189]. The AMPK pathway is known to be involved in the regulation of energy homeostasis via inhibiting de novo lipogenesis and adipogenesis [190]. Raspberry seed flour supplementation (100 mg EA/Kg BW) in a high sucrose diet mouse model has been used to demonstrate a reduction in hepatic endoplasmic reticulum stress, dyslipidemia, and adipose tissue inflammation, supporting the role of EA in preventing sugar toxicity [191]. Panchal and colleagues suggested EA supplementation (80 mg EA/kg BW) to be beneficial to improving the liver structure and function of HFD-induced male Wistar rats via blunting oxidative stress and inflammation. Administration of EA supplementation in the above study reduced the Metabolic Syndrome by regulating the protein levels of Nrf2, Nf-kB, and carnitine palmitoyl transferase-1 to their basal levels [108]. Further to this, EA has been proposed as a therapeutic nutrient for NAFLD. This was demonstrated when large lipid accumulation in HFD-induced liver disease was eliminated by punicalagin-enriched pomegranate extract (150 EA mg/kg/day) treatment in a rat model, where a marked reduction in liver triglyceride and cholesterol levels was observed [61]. Defatted walnut power extract contains many active polar substances including EA [192,193,194]. Ren and colleagues [195] have conducted a study investigating the anti-NAFLD effects of this walnut extract on an HFD-induced mouse model. The results showed that defatted walnut power extract reduced the expression of Nf-kB and the mitogen-activated protein kinases (MAPK) family, thus inhibiting inflammation in the liver during disease progression. This study also demonstrated that the administration of defatted walnut power extract improved the gut microbiota diversity disrupted by NAFLD [195]. A more recent study demonstrated that EA ameliorates high fructose-induced hyperuricemia and NAFLD through activation of C1q/tumour necrosis factor-related protein (CTRP3) and inhibition of ATP citrate lyase (ACL) in male albino rats [196]. Hyperuricemia occurs due to the damaged metabolism of uric acid [197] and is attributed as a risk factor for NAFLD [198,199,200].
New insights into research on EAs have focused on its microbial metabolites, the urolithins. According to Abdulrahman and colleagues [133], supplementation of urolithin A to HFD-fed Wistar rats, reduced bodyweight, serum levels of cholesterol and low-density lipoprotein (LDL), and increased levels of high-density lipoprotein (HDL). Moreover, this study demonstrated that urolithin A administration reduced the abundance of the specific microbiome related to weight gain, dysfunction of lipid metabolism, and impaired glucose metabolism, suggesting urolithin A has the potential to act as a therapeutic agent in obesity [133]. A study on both EA and urolithin A has revealed that in a high-fat/high-sucrose-fed mouse model, only EA demonstrated the capability to reduce proton leakage in primary hepatocytes, as opposed to urolithin A; thus, reporting its involvement in mitochondrial respiratory capacity during insulin resistance [201]. Mitochondrial proton leak can lead to impaired insulin signalling pathways, contributing to the development of insulin resistance, a key factor in NAFLD progression [44,202,203]. Intragastrical administration of urolithin A (50 or 100 mg/kg per day) improved hepatic steatosis induced by fructose consumption in a high-fructose-fed mouse model [204]. The results also stated that urolithin A inhibited lipogenesis while enhancing an increase in β-oxidation in the liver. Additionally, it promoted hepatic lipophagy through the AMPK/ULK1(Unc-51-like kinase 1) pathway. The AMPK/ULK1 pathway serves as a key regulator of lipid metabolism and autophagy within liver cells [205]. This suggests that AMPK/ULK1 regulates hepatic lipophagy when stimulated by urolithin A, thereby being a potential therapeutic for the treatment of NAFLD.
Xu and colleagues [206] in a recent study, used oral administration of urolithin C on a choline-deficient amino acid-defined high-fat-diet (CDAHFD) mouse model and demonstrated significantly improved liver index (weight of liver/body) and NAS score compared to the control and disease groups [206]. This study also revealed that the administration of urolithin C inhibited ferroptosis through activation of the hepatic AMP-activated protein kinase (AMPK) pathway [206]. Ferroptosis is a form of ion-dependent cell death and is recognised for its involvement in the pathogenesis of NAFLD. It is characterised by elevated oxidative stress and dysfunctional lipid metabolism [207,208]. Xu and colleagues also demonstrated that urolithin C ameliorated the permeability of the intestinal epithelial and increased the proportions of certain beneficial bacteria including Parabacteroides goldsteinii and Lactobacillus vaginalis, improving the dysbiosis caused by CDAHFD [206]. Another study conducted on Wistar rats on an HFD showed that both urolithins A and B improved characteristics associated with obesity, including weight gain, lipid accumulation, and oxidative stress [209]. The results of this study also revealed that both urolithins significantly downregulated the expression of liver X receptor (LXRα) and sterol regulatory element-binding protein-1c (SREBP1c) genes, which are involved in de novo lipogenesis. The authors showed that both urolithins A and B attenuate hepatic endoplasmic reticulum stress through the downregulation of unfolded protein responses [209].
The biological properties of EA have been investigated in several in vitro studies. One of the studies revealed that inhibiting HSC activation is highly sensitive to EA [210], suggesting its potential involvement in antifibrotic mechanisms. Furthermore, an in vitro study investigating the inhibitory effects of Phyllanthus emblica L. (Indian gooseberry) on hepatic steatosis and liver fibrosis identified EA as the main compound present in the water extract of P. emblica fruits [211]. In the study, a two-cell in vitro system to simulate NASH and hepatic fibrosis features, the authors used human hepatoblastoma HepG2 cells treated with a mixture of free fatty acids and the rat hepatic stellate HSC-T6 cells induced by leptin, respectively. The results showed the water extract reduced fat accumulation and ROS production through the modulation of the AMPK signalling pathway in HepG2 cells. They also demonstrated the potential for attenuating hepatic fibrosis in HSC-T6 cells and triggering mitochondrial apoptosis. Thus, indicating that EA has the potential to mitigate the progression of NAFLD [211].
Ellagic acid suppressed de novo lipogenesis by inhibiting the expression of sterol regulatory element-binding protein-1(SREBP-1)/fatty acid synthase (FASN) cascade and reducing steatosis in human hepatoma cell line through activation of AKT/mTORC1 (protein kinase B/mammalian target of rapamycin) in the liver [212]. Both SREBP-1 and FASN play a critical role in the increased production of fatty acids and exacerbating liver fat accumulation during NAFLD progression [213,214]. Dysregulation of the AKT/mTORC1 pathway can contribute to abnormal lipid metabolism, hepatocyte proliferation, and inflammation, ultimately promoting the development and progression [215,216]. Therefore, EA may hold therapeutic potential for managing NAFLD by targeting this pathway.
An in vitro study revealed that urolithin A promoted fatty acid breakdown, including both lipophagy and β-oxidation, via an AMPK-dependent pathway on fructose-treated HepG2 cells and primary hepatocytes. These in vitro experiments extended the findings from the high fructose-fed mouse model discussed above [204]. Additionally, a study conducted on oleic-acid-stimulated Alpha mouse liver 12 (AML12) cells revealed that urolithin C can activate the hepatic AMPK pathway to alleviate ferroptosis response in vivo but not in vitro [206]; thus, suggesting a crucial link between urolithin C and hepatic AMPK, likely through the gut-liver axis discussed earlier. Additionally, this study concluded that urolithin C was unable to reduce lipid accumulation or inhibit ferroptosis in vitro [206].
Consequently, in vitro findings suggest that both EA and urolithin possess the ability to regulate lipid metabolism during the progression of NAFLD. However, current data suggest that EA offers greater advantages compared to urolithin as it can inhibit both hepatic steatosis and hepatic fibrosis in vitro. Therefore, EA holds promise as a therapeutic agent for blunting the progression of the disease.
Despite a large amount of evidence being available in the form of pre-clinical studies, there are no clinical trials to date that have tested EA and its pharmacological properties on liver disease. A systematic review conducted by Gheflati and colleagues revealed that, even though many studies consider pomegranate as a tool to manage weight loss, there was no significant effect of pomegranate on body weight, BMI, and body fat percentage [217]. However, EA and pomegranate juice (naturally high in EA) have been evaluated in Phase I, II, and III clinical trials, primarily focusing on anticancer properties [218,219,220] and skin hyperpigmentation [221,222]. Even though there is a plethora of research on the potential hepatoprotective properties of EA, such as antioxidant and anti-inflammatory effects, most of these have not focused on Metabolic Syndrome, NAFLD, or diabetes.

7. Future Perspective of EA as a Pharmacological Therapy

EA stands out as a remarkable polyphenolic compound, possessing a wide range of pharmacological properties that hold promise in treating various chronic diseases, including NAFLD/NASH. Due to its multifaceted biological effects, edible plants containing EA and their derivatives are recognised as valuable functional foods for enhancing human health. Moreover, there is evidence suggesting that EA, when combined with other antioxidant nutraceuticals, exhibits a synergistic therapeutic effect, making it a potential candidate for combination therapy [113]. Although clinical trials investigating EA’s effects on NAFLD/NASH are pending, the pharmaceutical and cosmetic industries are already incorporating this polyphenolic compound into novel supplement preparations. Consequently, given the current widespread popularity of supplementation, it is important to consider incorporating EA as a dietary intervention for NAFLD/NASH. However, like many other polyphenols, the lack of comprehensive understanding regarding the underlying mechanisms governing its biological properties limits its capacity as a pharmacological agent in the market.

8. Conclusions

There is a great need for effective pharmacological treatments for NASH due to the severity, growing impact on the global health system, and more importantly, the absence of approved pharmacological treatments. EA exerts its hepatoprotective properties primarily through scavenging free radicals, modulating cytokine production, and regulating lipid metabolism. As an excellent antioxidant, EA acts against ROS and activates the NrF2 pathway to reduce oxidative stress to protect the liver. Remarkably, EA also suppresses Nf-kB and MAPK pathways, mitigating inflammation during NAFLD/NASH. The evidence also shows that EA can reduce both triglyceride and cholesterol levels, thus combating de novo lipogenesis, which is a significant risk factor in NASH progression. In vitro, findings suggest that EA has the capability to alleviate fibrosis. The primary microbial metabolite for EA, urolithin, has been shown to improve the gut microbiome in several mouse models of obesity. Specifically, urolithin A has been shown to lower LDL and increase HDL levels and is also involved in improving lipid metabolism through gene regulation, while urolithin C activates the hepatic AMPK pathway, thus counteracting the pathophysiology of NAFLD. There is an ongoing debate regarding the health benefits of EA and urolithins for NAFLD/NASH, but there is still a lack of understanding regarding their biological effect on the liver. Given the involvement of lipid metabolism, oxidative stress, inflammation, and insulin resistance in the pathogenesis of NASH, findings from this review suggest that EA may represent a potential food intervention for NASH, not only to limit but potentially reverse the pathological manifestations of NAFLD/NASH.

Author Contributions

Conceptualization, T.S. and R.L.; original draft preparation, T.S.; review and editing, T.S., A.S., L.B. and R.L.; supervision, L.B., R.L. and A.S.; funding, L.B. and R.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by an HDR scholarship of Edith Cowan University and Ricky R. Lareu, Curtin Medical School, Faculty of Health Sciences, Curtin University, WA, Australia.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Younossi, Z.; Tacke, F.; Arrese, M.; Chander Sharma, B.; Mostafa, I.; Bugianesi, E.; Wai-Sun Wong, V.; Yilmaz, Y.; George, J.; Fan, J.; et al. Global Perspectives on Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Hepatology 2019, 69, 2672–2682. [Google Scholar] [CrossRef] [PubMed]
  2. Younossi, Z.M.; Loomba, R.; Rinella, M.E.; Bugianesi, E.; Marchesini, G.; Neuschwander-Tetri, B.A.; Serfaty, L.; Negro, F.; Caldwell, S.H.; Ratziu, V. Current and future therapeutic regimens for nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology 2018, 68, 361–371. [Google Scholar] [CrossRef] [PubMed]
  3. Golabi, P.; Paik, J.M.; Eberly, K.; de Avila, L.; Alqahtani, S.A.; Younossi, Z.M. Causes of death in patients with Non-alcoholic Fatty Liver Disease (NAFLD), alcoholic liver disease and chronic viral Hepatitis B and C. Ann. Hepatol. 2022, 27, 100556. [Google Scholar] [CrossRef] [PubMed]
  4. Estes, C.; Razavi, H.; Loomba, R.; Younossi, Z.; Sanyal, A.J. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 2018, 67, 123–133. [Google Scholar] [CrossRef] [PubMed]
  5. Armandi, A.; Bugianesi, E. Natural history of NASH. Liver Int. 2021, 41, 78–82. [Google Scholar] [CrossRef] [PubMed]
  6. Brunt, E.M.; Kleiner, D.E.; Wilson, L.A.; Belt, P.; Neuschwander-Tetri, B.A.; Network, N.C.R. Nonalcoholic fatty liver disease (NAFLD) activity score and the histopathologic diagnosis in NAFLD: Distinct clinicopathologic meanings. Hepatology 2011, 53, 810–820. [Google Scholar] [CrossRef] [PubMed]
  7. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
  8. Tilg, H.; Adolph, T.E.; Moschen, A.R. Multiple parallel hits hypothesis in nonalcoholic fatty liver disease: Revisited after a decade. Hepatology 2021, 73, 833–842. [Google Scholar] [CrossRef] [PubMed]
  9. 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]
  10. Yang, Z.; Wang, L. Current, emerging, and potential therapies for non-alcoholic steatohepatitis. Front. Pharmacol. 2023, 14, 1152042. [Google Scholar] [CrossRef]
  11. Dai, L. Current NASH Pathogenesis and Diagnostic Techniques and Treatments. Highlights Sci. Eng. Technol. 2023, 36, 1532–1538. [Google Scholar] [CrossRef]
  12. Harrison, S.A.; Loomba, R.; Dubourg, J.; Ratziu, V.; Noureddin, M. Clinical Trial Landscape in NASH. Clin. Gastroenterol. Hepatol. 2023, 21, 2001–2014. [Google Scholar] [CrossRef] [PubMed]
  13. Ganz, M.; Bukong, T.N.; Csak, T.; Saha, B.; Park, J.K.; Ambade, A.; Kodys, K.; Szabo, G. Progression of non-alcoholic steatosis to steatohepatitis and fibrosis parallels cumulative accumulation of danger signals that promote inflammation and liver tumors in a high fat-cholesterol-sugar diet model in mice. J. Transl. Med. 2015, 13, 193. [Google Scholar] [CrossRef]
  14. Alfei, S.; Marengo, B.; Zuccari, G. Oxidative stress, antioxidant capabilities, and bioavailability: Ellagic acid or urolithins? Antioxidants 2020, 9, 707. [Google Scholar] [CrossRef]
  15. Cornelli, U. Antioxidant use in nutraceuticals. Clin. Dermatol. 2009, 27, 175–194. [Google Scholar] [CrossRef] [PubMed]
  16. Salomone, F.; Godos, J.; Zelber-Sagi, S. Natural antioxidants for non-alcoholic fatty liver disease: Molecular targets and clinical perspectives. Liver Int. 2016, 36, 5–20. [Google Scholar] [CrossRef] [PubMed]
  17. Kang, I.; Buckner, T.; Shay, N.F.; Gu, L.; Chung, S. Improvements in metabolic health with consumption of ellagic acid and subsequent conversion into urolithins: Evidence and mechanisms. Adv. Nutr. 2016, 7, 961–972. [Google Scholar] [CrossRef] [PubMed]
  18. Garcia-Nino, W.R.; Zazueta, C. Ellagic acid: Pharmacological activities and molecular mechanisms involved in liver protection. Pharmacol. Res. 2015, 97, 84–103. [Google Scholar] [CrossRef] [PubMed]
  19. Karimi, M.Y.; Fatemi, I.; Kalantari, H.; Mombeini, M.A.; Mehrzadi, S.; Goudarzi, M. Ellagic acid prevents oxidative stress, inflammation, and histopathological alterations in acrylamide-induced hepatotoxicity in wistar rats. J. Diet. Suppl. 2020, 17, 651–662. [Google Scholar] [CrossRef]
  20. Landete, J. Ellagitannins, ellagic acid and their derived metabolites: A review about source, metabolism, functions and health. Food Res. Int. 2011, 44, 1150–1160. [Google Scholar] [CrossRef]
  21. Aishwarya, V.; Solaipriya, S.; Sivaramakrishnan, V. Role of ellagic acid for the prevention and treatment of liver diseases. Phytother. Res. 2021, 35, 2925–2944. [Google Scholar] [CrossRef] [PubMed]
  22. Espín, J.C.; García-Conesa, M.T.; Tomás-Barberán, F.A. Nutraceuticals: Facts and fiction. Phytochemistry 2007, 68, 2986–3008. [Google Scholar] [CrossRef] [PubMed]
  23. Farrell, G.C.; Larter, C.Z. Nonalcoholic fatty liver disease: From steatosis to cirrhosis. Hepatology 2006, 43, S99–S112. [Google Scholar] [CrossRef] [PubMed]
  24. Day, C.; Saksena, S. Non-alcoholic steatohepatitis: Definitions and pathogenesis. J. Gastroenterol. Hepatol. 2002, 17, S377–S384. [Google Scholar] [CrossRef] [PubMed]
  25. Dowman, J.K.; Tomlinson, J.W.; Newsome, P.N. Pathogenesis of non-alcoholic fatty liver disease. QJM 2010, 103, 71–83. [Google Scholar] [CrossRef] [PubMed]
  26. Peverill, W.; Powell, L.W.; Skoien, R. Evolving Concepts in the Pathogenesis of NASH: Beyond Steatosis and Inflammation. Int. J. Mol. Sci. 2014, 15, 8591–8638. [Google Scholar] [CrossRef] [PubMed]
  27. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wong, V.W.-S.; Dufour, J.-F.; Schattenberg, J.M. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef] [PubMed]
  28. Méndez-Sánchez, N.; Bugianesi, E.; Gish, R.G.; Lammert, F.; Tilg, H.; Nguyen, M.H.; Sarin, S.K.; Fabrellas, N.; Zelber-Sagi, S.; Fan, J.-G. Global multi-stakeholder endorsement of the MAFLD definition. Lancet Gastroenterol. Hepatol. 2022, 7, 388–390. [Google Scholar] [CrossRef]
  29. Liu, J.; Ayada, I.; Zhang, X.; Wang, L.; Li, Y.; Wen, T.; Ma, Z.; Bruno, M.J.; de Knegt, R.J.; Cao, W.; et al. Estimating Global Prevalence of Metabolic Dysfunction-Associated Fatty Liver Disease in Overweight or Obese Adults. Clin. Gastroenterol. Hepatol. 2022, 20, e573–e582. [Google Scholar] [CrossRef]
  30. Eslam, M.; Sanyal, A.J.; George, J.; Sanyal, A.; Neuschwander-Tetri, B.; Tiribelli, C.; Kleiner, D.E.; Brunt, E.; Bugianesi, E.; Yki-Järvinen, H. MAFLD: A consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef]
  31. Zhou, J.; Sun, D.-Q.; Targher, G.; Byrne, C.D.; Lee, B.-w.; Hamaguchi, M.; Kim, S.U.; Hou, X.; Fadini, G.P.; Shimabukuro, M. Metabolic dysfunction-associated fatty liver disease increases risk of chronic kidney disease: A systematic review and meta-analysis. eGastroenterology 2023, 1, e100005. [Google Scholar] [CrossRef]
  32. Chan, K.E.; Koh, T.J.L.; Tang, A.S.P.; Quek, J.; Yong, J.N.; Tay, P.; Tan, D.J.H.; Lim, W.H.; Lin, S.Y.; Huang, D. Global prevalence and clinical characteristics of metabolic-associated fatty liver disease: A meta-analysis and systematic review of 10 739 607 individuals. J. Clin. Endocrinol. Metab. 2022, 107, 2691–2700. [Google Scholar] [CrossRef] [PubMed]
  33. Farrell, G.; Schattenberg, J.M.; Leclercq, I.; Yeh, M.M.; Goldin, R.; Teoh, N.; Schuppan, D. Mouse Models of Nonalcoholic Steatohepatitis: Toward Optimization of Their Relevance to Human Nonalcoholic Steatohepatitis. Hepatology 2019, 69, 2241–2257. [Google Scholar] [CrossRef]
  34. Guilherme, A.; Virbasius, J.V.; Puri, V.; Czech, M.P. Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes. Nat. Rev. Mol. Cell Biol. 2008, 9, 367–377. [Google Scholar] [CrossRef] [PubMed]
  35. Rosso, C.; Kazankov, K.; Younes, R.; Esmaili, S.; Marietti, M.; Sacco, M.; Carli, F.; Gaggini, M.; Salomone, F.; Møller, H.J. Crosstalk between adipose tissue insulin resistance and liver macrophages in non-alcoholic fatty liver disease. J. Hepatol. 2019, 71, 1012–1021. [Google Scholar] [CrossRef]
  36. Gehrke, N.; Schattenberg, J.M. Metabolic inflammation—A role for hepatic inflammatory pathways as drivers of comorbidities in nonalcoholic fatty liver disease? Gastroenterology 2020, 158, 1929–1947.e6. [Google Scholar] [CrossRef]
  37. Petrescu, M.; Vlaicu, S.I.; Ciumărnean, L.; Milaciu, M.V.; Mărginean, C.; Florea, M.; Vesa, Ș.C.; Popa, M. Chronic inflammation—A link between nonalcoholic fatty liver disease (NAFLD) and dysfunctional adipose tissue. Medicina 2022, 58, 641. [Google Scholar] [CrossRef] [PubMed]
  38. Stojsavljević, S.; Palčić, M.G.; Jukić, L.V.; Duvnjak, L.S.; Duvnjak, M. Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World J. Gastroenterol. WJG 2014, 20, 18070. [Google Scholar] [CrossRef]
  39. Tsochatzis, E.A.; Papatheodoridis, G.V.; Archimandritis, A.J. Adipokines in nonalcoholic steatohepatitis: From pathogenesis to implications in diagnosis and therapy. Mediat. Inflamm. 2009, 2009, 831670. [Google Scholar] [CrossRef]
  40. Das, S.K.; Balakrishnan, V. Role of cytokines in the pathogenesis of non-alcoholic fatty liver disease. Indian J. Clin. Biochem. 2011, 26, 202–209. [Google Scholar] [CrossRef]
  41. Alisi, A.; Carpino, G.; Oliveira, F.L.; Panera, N.; Nobili, V.; Gaudio, E. The Role of Tissue Macrophage-Mediated Inflammation on NAFLD Pathogenesis and Its Clinical Implications. Mediat. Inflamm. 2017, 2017, 8162421. [Google Scholar] [CrossRef] [PubMed]
  42. Diehl, A.; Li, Z.; Lin, H.; Yang, S. Cytokines and the pathogenesis of non-alcoholic steatohepatitis. Gut 2005, 54, 303. [Google Scholar] [CrossRef] [PubMed]
  43. Niederreiter, L.; Tilg, H. Cytokines and fatty liver diseases. Liver Res. 2018, 2, 14–20. [Google Scholar] [CrossRef]
  44. Mansouri, A.; Gattolliat, C.H.; Asselah, T. Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology 2018, 155, 629–647. [Google Scholar] [CrossRef] [PubMed]
  45. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef] [PubMed]
  46. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef] [PubMed]
  47. Safari, Z.; Gérard, P. The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cell. Mol. Life Sci. 2019, 76, 1541–1558. [Google Scholar] [CrossRef] [PubMed]
  48. Hildebrandt, M.A.; Hoffmann, C.; Sherrill–Mix, S.A.; Keilbaugh, S.A.; Hamady, M.; Chen, Y.Y.; Knight, R.; Ahima, R.S.; Bushman, F.; Wu, G.D. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 2009, 137, 1716–1724.e2. [Google Scholar] [CrossRef] [PubMed]
  49. Ogawa, Y.; Imajo, K.; Honda, Y.; Kessoku, T.; Tomeno, W.; Kato, S.; Fujita, K.; Yoneda, M.; Saito, S.; Saigusa, Y. Palmitate-induced lipotoxicity is crucial for the pathogenesis of nonalcoholic fatty liver disease in cooperation with gut-derived endotoxin. Sci. Rep. 2018, 8, 11365. [Google Scholar] [CrossRef]
  50. Yoo, J.Y.; Groer, M.; Dutra, S.V.O.; Sarkar, A.; McSkimming, D.I. Gut microbiota and immune system interactions. Microorganisms 2020, 8, 1587. [Google Scholar] [CrossRef]
  51. Liu, J.-P.; Zou, W.-L.; Chen, S.-J.; Wei, H.-Y.; Yin, Y.-N.; Zou, Y.-Y.; Lu, F.-G. Effects of different diets on intestinal microbiota and nonalcoholic fatty liver disease development. World J. Gastroenterol. 2016, 22, 7353. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, B.; Jiang, X.; Cao, M.; Ge, J.; Bao, Q.; Tang, L.; Chen, Y.; Li, L. Altered fecal microbiota correlates with liver biochemistry in nonobese patients with non-alcoholic fatty liver disease. Sci. Rep. 2016, 6, 32002. [Google Scholar] [CrossRef] [PubMed]
  53. Jiang, C.; Xie, C.; Li, F.; Zhang, L.; Nichols, R.G.; Krausz, K.W.; Cai, J.; Qi, Y.; Fang, Z.-Z.; Takahashi, S. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Investig. 2015, 125, 386–402. [Google Scholar] [CrossRef] [PubMed]
  54. Wahlström, A.; Sayin, S.I.; Marschall, H.-U.; Bäckhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed]
  55. Nimer, N.; Choucair, I.; Wang, Z.; Nemet, I.; Li, L.; Gukasyan, J.; Weeks, T.L.; Alkhouri, N.; Zein, N.; Tang, W.W. Bile acids profile, histopathological indices and genetic variants for non-alcoholic fatty liver disease progression. Metabolism 2021, 116, 154457. [Google Scholar] [CrossRef] [PubMed]
  56. Perez, M.J.; Briz, O. Bile-acid-induced cell injury and protection. World J. Gastroenterol. WJG 2009, 15, 1677. [Google Scholar] [CrossRef] [PubMed]
  57. Takaki, A.; Kawai, D.; Yamamoto, K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int. J. Mol. Sci. 2013, 14, 20704–20728. [Google Scholar] [CrossRef] [PubMed]
  58. Gonzalez, A.; Huerta-Salgado, C.; Orozco-Aguilar, J.; Aguirre, F.; Tacchi, F.; Simon, F.; Cabello-Verrugio, C. Role of Oxidative Stress in Hepatic and Extrahepatic Dysfunctions during Nonalcoholic Fatty Liver Disease (NAFLD). Oxidative Med. Cell. Longev. 2020, 2020, 1617805. [Google Scholar] [CrossRef] [PubMed]
  59. Cichoż-Lach, H.; Michalak, A. Oxidative stress as a crucial factor in liver diseases. World J. Gastroenterol. WJG 2014, 20, 8082. [Google Scholar] [CrossRef]
  60. Lebeaupin, C.; Vallée, D.; Hazari, Y.; Hetz, C.; Chevet, E.; Bailly-Maitre, B. Endoplasmic reticulum stress signalling and the pathogenesis of non-alcoholic fatty liver disease. J. Hepatol. 2018, 69, 927–947. [Google Scholar] [CrossRef]
  61. Zou, X.; Yan, C.; Shi, Y.; Cao, K.; Xu, J.; Wang, X.; Chen, C.; Luo, C.; Li, Y.; Gao, J.; et al. Mitochondrial dysfunction in obesity-associated nonalcoholic fatty liver disease: The protective effects of pomegranate with its active component punicalagin. Antioxid. Redox Signal. 2014, 21, 1557–1570. [Google Scholar] [CrossRef] [PubMed]
  62. Sakaida, I.; Okita, K. The role of oxidative stress in NASH and fatty liver model. Hepatol. Res. 2005, 33, 128–131. [Google Scholar] [CrossRef] [PubMed]
  63. Borrelli, A.; Bonelli, P.; Tuccillo, F.M.; Goldfine, I.D.; Evans, J.L.; Buonaguro, F.M.; Mancini, A. Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol. 2018, 15, 467–479. [Google Scholar] [CrossRef] [PubMed]
  64. Sanyal, A.J.; Campbell–Sargent, C.; Mirshahi, F.; Rizzo, W.B.; Contos, M.J.; Sterling, R.K.; Luketic, V.A.; Shiffman, M.L.; Clore, J.N. Nonalcoholic steatohepatitis: Association of insulin resistance and mitochondrial abnormalities. Gastroenterology 2001, 120, 1183–1192. [Google Scholar] [CrossRef] [PubMed]
  65. Berardo, C.; Di Pasqua, L.G.; Cagna, M.; Richelmi, P.; Vairetti, M.; Ferrigno, A. Nonalcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis: Current Issues and Future Perspectives in Preclinical and Clinical Research. Int. J. Mol. Sci. 2020, 21, 9646. [Google Scholar] [CrossRef]
  66. Postic, C.; Girard, J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: Lessons from genetically engineered mice. J. Clin. Investig. 2008, 118, 829–838. [Google Scholar] [CrossRef] [PubMed]
  67. Köhn-Gaone, J.; Gogoi-Tiwari, J.; Ramm, G.A.; Olynyk, J.K.; Tirnitz-Parker, J.E. The role of liver progenitor cells during liver regeneration, fibrogenesis, and carcinogenesis. Am. J. Physiol.-Gastrointest. Liver Physiol. 2016, 310, G143–G154. [Google Scholar] [CrossRef]
  68. Tirnitz-Parker, J.E.; Yeoh, G.C.; Olynyk, J.K. Liver progenitor cells, cancer stem cells and hepatocellular carcinoma. In Liver Regeneration; InTech: Rijeka, Croatia, 2012; p. 252. [Google Scholar]
  69. Negi, C.K.; Babica, P.; Bajard, L.; Bienertova-Vasku, J.; Tarantino, G. Insights into the molecular targets and emerging pharmacotherapeutic interventions for nonalcoholic fatty liver disease. Metabolism 2022, 126, 154925. [Google Scholar] [CrossRef] [PubMed]
  70. Polyzos, S.A.; Kang, E.S.; Boutari, C.; Rhee, E.-J.; Mantzoros, C.S. Current and emerging pharmacological options for the treatment of nonalcoholic steatohepatitis. Metabolism 2020, 111, 154203. [Google Scholar] [CrossRef]
  71. Peng, C.; Stewart, A.G.; Woodman, O.L.; Ritchie, R.H.; Qin, C.X. Non-Alcoholic Steatohepatitis: A Review of Its Mechanism, Models and Medical Treatments. Front. Pharmacol. 2020, 11, 603926. [Google Scholar] [CrossRef]
  72. Sumida, Y.; Yoneda, M. Current and future pharmacological therapies for NAFLD/NASH. J. Gastroenterol. 2018, 53, 362–376. [Google Scholar] [CrossRef] [PubMed]
  73. Elsheikh, E.; Younoszai, Z.; Otgonsuren, M.; Hunt, S.; Raybuck, B.; Younossi, Z.M. Markers of endothelial dysfunction in patients with non-alcoholic fatty liver disease and coronary artery disease. J. Gastroenterol. Hepatol. 2014, 29, 1528–1534. [Google Scholar] [CrossRef] [PubMed]
  74. Korinkova, L.; Prazienkova, V.; Cerna, L.; Karnosova, A.; Zelezna, B.; Kunes, J.; Maletinska, L. Pathophysiology of NAFLD and NASH in Experimental Models: The Role of Food Intake Regulating Peptides. Front. Endocrinol. 2020, 11, 597583. [Google Scholar] [CrossRef] [PubMed]
  75. Yoon, I.C.; Eun, J.R. Pharmacologic therapy for nonalcoholic steatohepatitis focusing on pathophysiology. Yeungnam Univ. J. Med. 2019, 36, 67–77. [Google Scholar] [CrossRef] [PubMed]
  76. Vancells Lujan, P.; Vinas Esmel, E.; Sacanella Meseguer, E. Overview of non-alcoholic fatty liver disease (NAFLD) and the role of sugary food consumption and other dietary components in its development. Nutrients 2021, 13, 1442. [Google Scholar] [CrossRef] [PubMed]
  77. Berná, G.; Romero-Gomez, M. The role of nutrition in non-alcoholic fatty liver disease: Pathophysiology and management. Liver Int. 2020, 40, 102–108. [Google Scholar] [CrossRef]
  78. Semmler, G.; Datz, C.; Reiberger, T.; Trauner, M. Diet and exercise in NAFLD/NASH: Beyond the obvious. Liver Int. 2021, 41, 2249–2268. [Google Scholar] [CrossRef] [PubMed]
  79. Perdomo, C.M.; Frühbeck, G.; Escalada, J. Impact of nutritional changes on nonalcoholic fatty liver disease. Nutrients 2019, 11, 677. [Google Scholar] [CrossRef]
  80. Bhoite, R.; Joshi, N.A.; Pratti, V.L.; Satyavrat, V. A Review on the Role of Low Glycemic Index Foods for Glycemic Control in Chronic Liver Disease. Food Nutr. Sci. 2023, 14, 258–276. [Google Scholar] [CrossRef]
  81. Parker, A.; Kim, Y. The effect of low glycemic index and glycemic load diets on hepatic fat mass, insulin resistance, and blood lipid panels in individuals with nonalcoholic fatty liver disease. Metab. Syndr. Relat. Disord. 2019, 17, 389–396. [Google Scholar] [CrossRef]
  82. Cheah, M.C.; McCullough, A.J.; Goh, G.B. Dietary manipulations for nonalcoholic fatty liver disease (NAFLD). In Bioactive Food as Dietary Interventions for Diabetes; Elsevier: Amsterdam, The Netherlands, 2019; pp. 69–88. [Google Scholar]
  83. Carpi, R.Z.; Barbalho, S.M.; Sloan, K.P.; Laurindo, L.F.; Gonzaga, H.F.; Grippa, P.C.; Zutin, T.L.M.; Girio, R.J.; Repetti, C.S.F.; Detregiachi, C.R.P. The effects of probiotics, prebiotics and synbiotics in non-alcoholic fat liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH): A systematic review. Int. J. Mol. Sci. 2022, 23, 8805. [Google Scholar] [CrossRef]
  84. Cicero, A.F.G.; Colletti, A.; Bellentani, S. Nutraceutical Approach to Non-Alcoholic Fatty Liver Disease (NAFLD): The Available Clinical Evidence. Nutrients 2018, 10, 1153. [Google Scholar] [CrossRef]
  85. Rives, C.; Fougerat, A.; Ellero-Simatos, S.; Loiseau, N.; Guillou, H.; Gamet-Payrastre, L.; Wahli, W. Oxidative stress in NAFLD: Role of nutrients and food contaminants. Biomolecules 2020, 10, 1702. [Google Scholar] [CrossRef]
  86. Moroshko, I.; Brennan, L.; O’Brien, P. Predictors of dropout in weight loss interventions: A systematic review of the literature. Obes. Rev. 2011, 12, 912–934. [Google Scholar] [CrossRef] [PubMed]
  87. Campbell, P.; Symonds, A.; Barritt, A.S.t. Therapy for Nonalcoholic Fatty Liver Disease: Current Options and Future Directions. Clin. Ther. 2021, 43, 500–517. [Google Scholar] [CrossRef]
  88. Harvey, B.E. NASH: Regulatory considerations for clinical drug development and US FDA approval. Acta Pharmacol. Sin. 2022, 43, 1210–1214. [Google Scholar] [CrossRef]
  89. Kothari, S.; Dhami-Shah, H.; Shah, S.R. Antidiabetic Drugs and Statins in Nonalcoholic Fatty Liver Disease. J. Clin. Exp. Hepatol. 2019, 9, 723–730. [Google Scholar] [CrossRef] [PubMed]
  90. Armstrong, M.J.; Hull, D.; Guo, K.; Barton, D.; Hazlehurst, J.M.; Gathercole, L.L.; Nasiri, M.; Yu, J.; Gough, S.C.; Newsome, P.N.; et al. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J. Hepatol. 2016, 64, 399–408. [Google Scholar] [CrossRef] [PubMed]
  91. Armstrong, M.J.; Barton, D.; Gaunt, P.; Hull, D.; Guo, K.; Stocken, D.; Gough, S.C.L.; Tomlinson, J.W.; Brown, R.M.; Hübscher, S.G.; et al. Liraglutide efficacy and action in non-alcoholic steatohepatitis (LEAN): Study protocol for a phase II multicentre, double-blinded, randomised, controlled trial. BMJ Open 2013, 3, e003995. [Google Scholar] [CrossRef]
  92. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; Abouda, G.; Aldersley, M.A.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016, 387, 679–690. [Google Scholar] [CrossRef]
  93. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.-S.; Harrison, S.A. A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef] [PubMed]
  94. Belfort, R.; Harrison, S.A.; Brown, K.; Darland, C.; Finch, J.; Hardies, J.; Balas, B.; Gastaldelli, A.; Tio, F.; Pulcini, J.; et al. A Placebo-Controlled Trial of Pioglitazone in Subjects with Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2006, 355, 2297–2307. [Google Scholar] [CrossRef] [PubMed]
  95. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  96. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [PubMed]
  97. Mahady, S.E.; Webster, A.C.; Walker, S.; Sanyal, A.; George, J. The role of thiazolidinediones in non-alcoholic steatohepatitis—A systematic review and meta analysis. J. Hepatol. 2011, 55, 1383–1390. [Google Scholar] [CrossRef] [PubMed]
  98. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N. Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef] [PubMed]
  99. Caldwell, S. NASH Therapy: Omega 3 supplementation, vitamin E, insulin sensitizers and statin drugs. Clin. Mol. Hepatol. 2017, 23, 103–108. [Google Scholar] [CrossRef] [PubMed]
  100. Singh, U.; Devaraj, S.; Jialal, I. Vitamin E, oxidative stress, and inflammation. Annu. Rev. Nutr. 2005, 25, 151–174. [Google Scholar] [CrossRef] [PubMed]
  101. Klein, E.A.; Thompson, I.M.; Tangen, C.M.; Crowley, J.J.; Lucia, M.S.; Goodman, P.J.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; et al. Vitamin E and the Risk of Prostate Cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556. [Google Scholar] [CrossRef]
  102. Lippman, S.M.; Klein, E.A.; Goodman, P.J.; Lucia, M.S.; Thompson, I.M.; Ford, L.G.; Parnes, H.L.; Minasian, L.M.; Gaziano, J.M.; Hartline, J.A.; et al. Effect of Selenium and Vitamin E on Risk of Prostate Cancer and Other Cancers: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2009, 301, 39–51. [Google Scholar] [CrossRef]
  103. Saremi, A.; Arora, R. Vitamin E and Cardiovascular Disease. Am. J. Ther. 2010, 17, e56–e65. [Google Scholar] [CrossRef] [PubMed]
  104. 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]
  105. Sinha, R.A.; Bruinstroop, E.; Singh, B.K.; Yen, P.M. Nonalcoholic fatty liver disease and hypercholesterolemia: Roles of thyroid hormones, metabolites, and agonists. Thyroid 2019, 29, 1173–1191. [Google Scholar] [CrossRef]
  106. Harrison, S.A.; Bashir, M.R.; Guy, C.D.; Zhou, R.; Moylan, C.A.; Frias, J.P.; Alkhouri, N.; Bansal, M.B.; Baum, S.; Neuschwander-Tetri, B.A. Resmetirom (MGL-3196) for the treatment of non-alcoholic steatohepatitis: A multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2019, 394, 2012–2024. [Google Scholar] [CrossRef] [PubMed]
  107. Harrison, S.A.; Taub, R.; Neff, G.W.; Lucas, K.J.; Labriola, D.; Moussa, S.E.; Alkhouri, N.; Bashir, M.R. Resmetirom for nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled phase 3 trial. Nat. Med. 2023, 29, 2919–2928. [Google Scholar] [CrossRef]
  108. Panchal, S.K.; Ward, L.; Brown, L. Ellagic acid attenuates high-carbohydrate, high-fat diet-induced metabolic syndrome in rats. Eur. J. Nutr. 2013, 52, 559–568. [Google Scholar] [CrossRef]
  109. Ríos, J.-L.; Giner, R.M.; Marín, M.; Recio, M.C. A pharmacological update of ellagic acid. Planta Medica 2018, 84, 1068–1093. [Google Scholar] [CrossRef]
  110. Rossi, M.; Erlebacher, J.; Zacharias, D.E.; Carrell, H.; Iannucci, B. The crystal and molecular structure of ellagic acid dihydrate: A dietary anti-cancer agent. Carcinogenesis 1991, 12, 2227–2232. [Google Scholar] [CrossRef]
  111. Clifford, M.N.; Scalbert, A. Ellagitannins–nature, occurrence and dietary burden. J. Sci. Food Agric. 2000, 80, 1118–1125. [Google Scholar] [CrossRef]
  112. Mansouri, M.T.; Farbood, Y.; Naghizadeh, B.; Shabani, S.; Mirshekar, M.A.; Sarkaki, A. Beneficial effects of ellagic acid against animal models of scopolamine-and diazepam-induced cognitive impairments. Pharm. Biol. 2016, 54, 1947–1953. [Google Scholar] [CrossRef]
  113. Altındağ, F.; Rağbetli, M.Ç.; Özdek, U.; Koyun, N.; Alhalboosi, J.K.I.; Elasan, S. Combined treatment of sinapic acid and ellagic acid attenuates hyperglycemia in streptozotocin-induced diabetic rats. Food Chem. Toxicol. 2021, 156, 112443. [Google Scholar] [CrossRef] [PubMed]
  114. Tomas-Barberan, F.A.; Andres-Lacueva, C. Polyphenols and health: Current state and progress. J. Agric. Food Chem. 2012, 60, 8773–8775. [Google Scholar] [CrossRef]
  115. Wang, W.; Wang, S.; Liu, Y.; Wang, X.; Nie, J.; Meng, X.; Zhang, Y. Ellagic acid: A dietary-derived phenolic compound for drug discovery in mild cognitive impairment. Front. Aging Neurosci. 2022, 14, 925855. [Google Scholar] [CrossRef] [PubMed]
  116. Lin, I.C.; Wu, J.-Y.; Fang, C.-Y.; Wang, S.-C.; Liu, Y.-W.; Ho, S.-T. Absorption and Metabolism of Urolithin A and Ellagic Acid in Mice and Their Cytotoxicity in Human Colorectal Cancer Cells. Evid.-Based Complement. Altern. Med. 2023, 2023, 8264716. [Google Scholar] [CrossRef]
  117. Cerdá, B.; Llorach, R.; Cerón, J.J.; Espín, J.C.; Tomás-Barberán, F.A. Evaluation of the bioavailability and metabolism in the rat of punicalagin, an antioxidant polyphenol from pomegranate juice. Eur. J. Nutr. 2003, 42, 18–28. [Google Scholar] [CrossRef] [PubMed]
  118. 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]
  119. Tomás-Barberán, F.A.; García-Villalba, R.; González-Sarrías, A.; Selma, M.V.; Espín, J.C. Ellagic Acid Metabolism by Human Gut Microbiota: Consistent Observation of Three Urolithin Phenotypes in Intervention Trials, Independent of Food Source, Age, and Health Status. J. Agric. Food Chem. 2014, 62, 6535–6538. [Google Scholar] [CrossRef] [PubMed]
  120. Cerdá, B.; Tomás-Barberán, F.A.; Espín, J.C. Metabolism of antioxidant and chemopreventive ellagitannins from strawberries, raspberries, walnuts, and oak-aged wine in humans: Identification of biomarkers and individual variability. J. Agric. Food Chem. 2005, 53, 227–235. [Google Scholar] [CrossRef] [PubMed]
  121. Larrosa, M.; García-Conesa, M.T.; Espín, J.C.; Tomás-Barberán, F.A. Ellagitannins, ellagic acid and vascular health. Mol. Asp. Med. 2010, 31, 513–539. [Google Scholar] [CrossRef]
  122. Karimi, E.; Ghorbani Nohooji, M.; Habibi, M.; Ebrahimi, M.; Mehrafarin, A.; Khalighi-Sigaroodi, F. Antioxidant potential assessment of phenolic and flavonoid rich fractions of Clematis orientalis and Clematis ispahanica (Ranunculaceae). Nat. Prod. Res. 2018, 32, 1991–1995. [Google Scholar] [CrossRef]
  123. Daniel, E.M.; Krupnick, A.S.; Heur, Y.-H.; Blinzler, J.A.; Nims, R.W.; Stoner, G.D. Extraction, stability, and quantitation of ellagic acid in various fruits and nuts. J. Food Compos. Anal. 1989, 2, 338–349. [Google Scholar] [CrossRef]
  124. Shendge, A.K.; Basu, T.; Chaudhuri, D.; Panja, S.; Mandal, N. In vitro antioxidant and antiproliferative activities of various solvent fractions from Clerodendrum viscosum leaves. Pharmacogn. Mag. 2017, 13, S344. [Google Scholar] [PubMed]
  125. 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]
  126. Larrosa, M.; González-Sarrías, A.; García-Conesa, M.T.; Tomás-Barberán, F.A.; Espín, J.C. Urolithins, ellagic acid-derived metabolites produced by human colonic microflora, exhibit estrogenic and antiestrogenic activities. J. Agric. Food Chem. 2006, 54, 1611–1620. [Google Scholar] [CrossRef] [PubMed]
  127. Gu, J.; Thomas-Ahner, J.M.; Riedl, K.M.; Bailey, M.T.; Vodovotz, Y.; Schwartz, S.J.; Clinton, S.K. Dietary black raspberries impact the colonic microbiome and phytochemical metabolites in mice. Mol. Nutr. Food Res. 2019, 63, 1800636. [Google Scholar] [CrossRef]
  128. 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] [PubMed]
  129. Tomás-Barberán, F.A.; González-Sarrías, A.; García-Villalba, R.; Núñez-Sánchez, M.A.; Selma, M.V.; García-Conesa, M.T.; Espín, J.C. Urolithins, the rescue of “old” metabolites to understand a “new” concept: Metabotypes as a nexus among phenolic metabolism, microbiota dysbiosis, and host health status. Mol. Nutr. Food Res. 2017, 61, 1500901. [Google Scholar] [CrossRef] [PubMed]
  130. García-Villalba, R.; Selma, M.V.; Espín, J.C.; Tomás-Barberán, F.A. Identification of novel urolithin metabolites in human feces and urine after the intake of a pomegranate extract. J. Agric. Food Chem. 2019, 67, 11099–11107. [Google Scholar] [CrossRef]
  131. Cortés-Martín, A.; García-Villalba, R.; González-Sarrías, A.; Romo-Vaquero, M.; Loria-Kohen, V.; Ramírez-de-Molina, A.; Tomás-Barberán, F.; Selma, M.; Espín, J. The gut microbiota urolithin metabotypes revisited: The human metabolism of ellagic acid is mainly determined by aging. Food Funct. 2018, 9, 4100–4106. [Google Scholar] [CrossRef]
  132. Selma, M.V.; Tomás-Barberán, F.A.; Beltrán, D.; García-Villalba, R.; Espín, J.C. Gordonibacter urolithinfaciens sp. nov., a urolithin-producing bacterium isolated from the human gut. Int. J. Syst. Evol. Microbiol. 2014, 64, 2346–2352. [Google Scholar] [CrossRef]
  133. Abdulrahman, A.O.; Alzubaidi, M.Y.; Nadeem, M.S.; Khan, J.A.; Rather, I.A.; Khan, M.I. The Utilization of Urolithin A—A Natural Polyphenol Metabolite of Ellagitannins as a Modulator of the Gut Microbiota for Its Potential Use in Obesity Therapy. Proceedings 2021, 79, 12. [Google Scholar]
  134. Zhang, M.; Cui, S.; Mao, B.; Zhang, Q.; Zhao, J.; Zhang, H.; Tang, X.; Chen, W. Ellagic acid and intestinal microflora metabolite urolithin A: A review on its sources, metabolic distribution, health benefits, and biotransformation. Crit. Rev. Food Sci. Nutr. 2023, 63, 6900–6922. [Google Scholar] [CrossRef]
  135. Beltrán, D.; Romo-Vaquero, M.; Espín, J.C.; Tomás-Barberán, F.A.; Selma, M.V. Ellagibacter isourolithinifaciens gen. nov., sp. nov., a new member of the family Eggerthellaceae, isolated from human gut. Int. J. Syst. Evol. Microbiol. 2018, 68, 1707–1712. [Google Scholar] [CrossRef] [PubMed]
  136. 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]
  137. Hamad, A.-W.R.; Al-Momani, W.M.; Janakat, S.; Oran, S.A. Bioavailability of ellagic acid after single dose administration using HPLC. Pak. J. Nutr. 2009, 8, 1661–1664. [Google Scholar] [CrossRef]
  138. Long, J.; Guo, Y.; Yang, J.; Henning, S.M.; Lee, R.-P.; Rasmussen, A.; Zhang, L.; Lu, Q.-Y.; Heber, D.; Li, Z. Bioavailability and bioactivity of free ellagic acid compared to pomegranate juice. Food Funct. 2019, 10, 6582–6588. [Google Scholar] [CrossRef]
  139. Tomás-Barberan, F.A.; Espín, J.C.; García-Conesa, M.T. Bioavailability and metabolism of ellagic acid and ellagitannins. In Chemistry and Biology of Ellagitannins: An Underestimated Class of Bioactive Plant Polyphenols; World Scientific: Singapore, 2009; pp. 273–297. [Google Scholar]
  140. Seeram, N.P.; Zhang, Y.; McKeever, R.; Henning, S.M.; Lee, R.-p.; Suchard, M.A.; Li, Z.; Chen, S.; Thames, G.; Zerlin, A. Pomegranate juice and extracts provide similar levels of plasma and urinary ellagitannin metabolites in human subjects. J. Med. Food 2008, 11, 390–394. [Google Scholar] [CrossRef] [PubMed]
  141. 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] [PubMed]
  142. Ren, S.-m.; Zhang, Q.-z.; Jiang, M.; Chen, M.-l.; Xu, X.-j.; Wang, D.-m.; Pan, Y.-n.; Liu, X.-q. Systematic characterization of the metabolites of defatted walnut powder extract in vivo and screening of the mechanisms against NAFLD by UPLC-Q-Exactive Orbitrap MS combined with network pharmacology. J. Ethnopharmacol. 2022, 285, 114870. [Google Scholar] [CrossRef]
  143. Stoner, G.D.; Sardo, C.; Apseloff, G.; Mullet, D.; Wargo, W.; Pound, V.; Singh, A.; Sanders, J.; Aziz, R.; Casto, B. Pharmacokinetics of anthocyanins and ellagic acid in healthy volunteers fed freeze-dried black raspberries daily for 7 days. J. Clin. Pharmacol. 2005, 45, 1153–1164. [Google Scholar] [CrossRef]
  144. Özkaya, A.; Celik, S.; Yüce, A.; Şahin, Z.; Yilmaz, Ö. The effects of ellagic acid on some biochemical parameters in the liver of rats against oxidative stress induced by aluminum. Kafkas Univ. Vet. Fak. Derg. 2010, 16, 263–268. [Google Scholar]
  145. Zeb, A. Ellagic acid in suppressing in vivo and in vitro oxidative stresses. Mol. Cell. Biochem. 2018, 448, 27–41. [Google Scholar] [CrossRef] [PubMed]
  146. Salem, A.M.; Mohammaden, T.F.; Ali, M.A.; Mohamed, E.A.; Hasan, H.F. Ellagic and ferulic acids alleviate gamma radiation and aluminium chloride-induced oxidative damage. Life Sci. 2016, 160, 2–11. [Google Scholar] [CrossRef] [PubMed]
  147. Iino, T.; Nakahara, K.; Miki, W.; Kiso, Y.; Ogawa, Y.; Kato, S.; Takeuchi, K. Less damaging effect of whisky in rat stomachs in comparison with pure ethanol. Digestion 2001, 64, 214–221. [Google Scholar] [CrossRef] [PubMed]
  148. Iino, T.; Tashima, K.; Umeda, M.; Ogawa, Y.; Takeeda, M.; Takata, K.; Takeuchi, K. Effect of ellagic acid on gastric damage induced in ischemic rat stomachs following ammonia or reperfusion. Life Sci. 2002, 70, 1139–1150. [Google Scholar] [CrossRef] [PubMed]
  149. Priyadarsini, K.I.; Khopde, S.M.; Kumar, S.S.; Mohan, H. Free radical studies of ellagic acid, a natural phenolic antioxidant. J. Agric. Food Chem. 2002, 50, 2200–2206. [Google Scholar] [CrossRef]
  150. Hassoun, E.; Walter, A.; Alsharif, N.; Stohs, S. Modulation of TCDD-induced fetotoxicity and oxidative stress in embryonic and placental tissues of C57BL/6J mice by vitamin E succinate and ellagic acid. Toxicology 1997, 124, 27–37. [Google Scholar] [CrossRef] [PubMed]
  151. Kilic, I.; Yeşiloğlu, Y.; Bayrak, Y. Spectroscopic studies on the antioxidant activity of ellagic acid. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2014, 130, 447–452. [Google Scholar] [CrossRef] [PubMed]
  152. Yu, Y.-M.; Chang, W.-C.; Wu, C.-H.; Chiang, S.-Y. Reduction of oxidative stress and apoptosis in hyperlipidemic rabbits by ellagic acid. J. Nutr. Biochem. 2005, 16, 675–681. [Google Scholar] [CrossRef]
  153. Craft, B.D.; Kerrihard, A.L.; Amarowicz, R.; Pegg, R.B. Phenol-based antioxidants and the in vitro methods used for their assessment. Compr. Rev. Food Sci. Food Saf. 2012, 11, 148–173. [Google Scholar] [CrossRef]
  154. Mira, L.; Tereza Fernandez, M.; Santos, M.; Rocha, R.; Helena Florêncio, M.; Jennings, K.R. Interactions of flavonoids with iron and copper ions: A mechanism for their antioxidant activity. Free Radic. Res. 2002, 36, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  155. Ahmed, S.; Rahman, A.; Saleem, M.; Athar, M.; Sultana, S. Ellagic acid ameliorates nickel induced biochemical alterations: Diminution of oxidative stress. Hum. Exp. Toxicol. 1999, 18, 691–698. [Google Scholar] [CrossRef] [PubMed]
  156. Saha, P.; Yeoh, B.S.; Singh, R.; Chandrasekar, B.; Vemula, P.K.; Haribabu, B.; Vijay-Kumar, M.; Jala, V.R. Gut microbiota conversion of dietary ellagic acid into bioactive phytoceutical urolithin A inhibits heme peroxidases. PLoS ONE 2016, 11, e0156811. [Google Scholar] [CrossRef] [PubMed]
  157. Galano, A.; Francisco Marquez, M.; Pérez-González, A. Ellagic acid: An unusually versatile protector against oxidative stress. Chem. Res. Toxicol. 2014, 27, 904–918. [Google Scholar] [CrossRef] [PubMed]
  158. Srinivasan, P.; Vadhanam, M.V.; Arif, J.M.; Gupta, R.C. A rapid screening assay for antioxidant potential of natural and synthetic agents in vitro. Int. J. Oncol. 2002, 20, 983–986. [Google Scholar] [CrossRef] [PubMed]
  159. Böyük, A.; Önder, A.; Kapan, M.; Gümüş, M.; Fιrat, U.; Başaralι, M.K.; Alp, H. Ellagic acid ameliorates lung injury after intestinal ischemia-reperfusion. Pharmacogn. Mag. 2011, 7, 224. [Google Scholar] [CrossRef] [PubMed]
  160. Thulstrup, P.W.; Thormann, T.; Spanget-Larsen, J.; Bisgaard, H.C. Interaction between ellagic acid and calf thymus DNA studied with flow linear dichroism UV–VIS spectroscopy. Biochem. Biophys. Res. Commun. 1999, 265, 416–421. [Google Scholar] [CrossRef] [PubMed]
  161. Teel, R.W.; Martin, R.M.; Allahyari, R. Ellagic acid metabolism and binding to DNA in organ explant cultures of the rat. Cancer Lett. 1987, 36, 203–211. [Google Scholar] [CrossRef]
  162. Spencer, W.A.; Jeyabalan, J.; Kichambre, S.; Gupta, R.C. Oxidatively generated DNA damage after Cu (II) catalysis of dopamine and related catecholamine neurotransmitters and neurotoxins: Role of reactive oxygen species. Free Radic. Biol. Med. 2011, 50, 139–147. [Google Scholar] [CrossRef]
  163. Li, W.; Kong, A.N. Molecular mechanisms of Nrf2-mediated antioxidant response. Mol. Carcinog. 2009, 48, 91–104. [Google Scholar] [CrossRef]
  164. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed]
  165. Ding, Y.; Zhang, B.; Zhou, K.; Chen, M.; Wang, M.; Jia, Y.; Song, Y.; Li, Y.; Wen, A. Dietary ellagic acid improves oxidant-induced endothelial dysfunction and atherosclerosis: Role of Nrf2 activation. Int. J. Cardiol. 2014, 175, 508–514. [Google Scholar] [CrossRef] [PubMed]
  166. Baek, B.; Lee, S.H.; Kim, K.; Lim, H.-W.; Lim, C.-J. Ellagic acid plays a protective role against UV-B-induced oxidative stress by up-regulating antioxidant components in human dermal fibroblasts. KJPP 2016, 20, 269–277. [Google Scholar] [CrossRef]
  167. Gu, L.; Deng, W.-S.; Liu, Y.; Jiang, C.-H.; Sun, L.-C.; Sun, X.-F.; Xu, Q.; Zhou, H. Ellagic acid protects Lipopolysaccharide/D-galactosamine-induced acute hepatic injury in mice. Int. Immunopharmacol. 2014, 22, 341–345. [Google Scholar] [CrossRef]
  168. Mishra, S.; Vinayak, M. Ellagic acid inhibits PKC signaling by improving antioxidant defense system in murine T cell lymphoma. Mol. Biol. Rep. 2014, 41, 4187–4197. [Google Scholar] [CrossRef]
  169. Biswas, S.K. Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxidative Med. Cell. Longev. 2016, 2016, 5698931. [Google Scholar] [CrossRef] [PubMed]
  170. Ziccardi, P.; Nappo, F.; Giugliano, G.; Esposito, K.; Marfella, R.; Cioffi, M.; D’Andrea, F.; Molinari, A.M.; Giugliano, D. Reduction of inflammatory cytokine concentrations and improvement of endothelial functions in obese women after weight loss over one year. Circulation 2002, 105, 804–809. [Google Scholar] [CrossRef]
  171. Le Marchand-Brustel, Y.; Gual, P.; Gremeaux, T.; Gonzalez, T.; Barres, R.; Tanti, J.-F. Fatty acid-induced insulin resistance: Role of insulin receptor substrate 1 serine phosphorylation in the retroregulation of insulin signalling. Biochem. Soc. Trans. 2003, 31, 1152–1156. [Google Scholar] [CrossRef]
  172. Lopez-Parra, V.; Mallavia, B.; Egido, J.; Gomez-Guerrero, C. Immunoinflammation in diabetic nephropathy: Molecular mechanisms and therapeutic options. In Diabetic Nephropathy; InTech: Rijeka, Croatia, 2012; pp. 127–146. [Google Scholar]
  173. Kuhad, A.; Chopra, K. Attenuation of diabetic nephropathy by tocotrienol: Involvement of NFkB signaling pathway. Life Sci. 2009, 84, 296–301. [Google Scholar] [CrossRef]
  174. Ahad, A.; Ganai, A.A.; Mujeeb, M.; Siddiqui, W.A. Ellagic acid, an NF-κB inhibitor, ameliorates renal function in experimental diabetic nephropathy. Chem.-Biol. Interact. 2014, 219, 64–75. [Google Scholar] [CrossRef]
  175. Chun, K.-S.; Cha, H.-H.; Shin, J.-W.; Na, H.-K.; Park, K.-K.; Chung, W.-Y.; Surh, Y.-J. Nitric oxide induces expression of cyclooxygenase-2 in mouse skin through activation of NF-κB. Carcinogenesis 2004, 25, 445–454. [Google Scholar] [CrossRef] [PubMed]
  176. El-Shitany, N.A.; El-Bastawissy, E.A.; El-desoky, K. Ellagic acid protects against carrageenan-induced acute inflammation through inhibition of nuclear factor kappa B, inducible cyclooxygenase and proinflammatory cytokines and enhancement of interleukin-10 via an antioxidant mechanism. Int. Immunopharmacol. 2014, 19, 290–299. [Google Scholar] [CrossRef] [PubMed]
  177. Romier, B.; Van De Walle, J.; During, A.; Larondelle, Y.; Schneider, Y.-J. Modulation of signalling nuclear factor-κB activation pathway by polyphenols in human intestinal Caco-2 cells. Br. J. Nutr. 2008, 100, 542–551. [Google Scholar] [CrossRef] [PubMed]
  178. 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]
  179. Sarkar, S.; Siddiqui, A.A.; Mazumder, S.; De, R.; Saha, S.J.; Banerjee, C.; Iqbal, M.S.; Adhikari, S.; Alam, A.; Roy, S. Ellagic acid, a dietary polyphenol, inhibits tautomerase activity of human macrophage migration inhibitory factor and its pro-inflammatory responses in human peripheral blood mononuclear cells. J. Agric. Food Chem. 2015, 63, 4988–4998. [Google Scholar] [CrossRef] [PubMed]
  180. Winand, J.; Schneider, Y.-J. The anti-inflammatory effect of a pomegranate husk extract on inflamed adipocytes and macrophages cultivated independently, but not on the inflammatory vicious cycle between adipocytes and macrophages. Food Funct. 2014, 5, 310–318. [Google Scholar] [CrossRef]
  181. Seo, C.-S.; Jeong, S.-J.; Yoo, S.-R.; Lee, N.-R.; Shin, H.-K. Quantitative analysis and in vitro anti-inflammatory effects of gallic acid, ellagic acid, and quercetin from Radix Sanguisorbae. Pharmacogn. Mag. 2016, 12, 104. [Google Scholar] [PubMed]
  182. Steppan, C.M.; Bailey, S.T.; Bhat, S.; Brown, E.J.; Banerjee, R.R.; Wright, C.M.; Patel, H.R.; Ahima, R.S.; Lazar, M.A. The hormone resistin links obesity to diabetes. Nature 2001, 409, 307–312. [Google Scholar] [CrossRef] [PubMed]
  183. Banerjee, R.R.; Rangwala, S.M.; Shapiro, J.S.; Rich, A.S.; Rhoades, B.; Qi, Y.; Wang, J.; Rajala, M.W.; Pocai, A.; Scherer, P.E. Regulation of fasted blood glucose by resistin. Science 2004, 303, 1195–1198. [Google Scholar] [CrossRef]
  184. Makino-Wakagi, Y.; Yoshimura, Y.; Uzawa, Y.; Zaima, N.; Moriyama, T.; Kawamura, Y. Ellagic acid in pomegranate suppresses resistin secretion by a novel regulatory mechanism involving the degradation of intracellular resistin protein in adipocytes. Biochem. Biophys. Res. Commun. 2012, 417, 880–885. [Google Scholar] [CrossRef]
  185. 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] [PubMed]
  186. Sharifi-Rad, J.; Quispe, C.; Castillo, C.M.S.; Caroca, R.; Lazo-Vélez, M.A.; Antonyak, H.; Polishchuk, A.; Lysiuk, R.; Oliinyk, P.; De Masi, L.; et al. Ellagic Acid: A Review on Its Natural Sources, Chemical Stability, and Therapeutic Potential. Oxidative Med. Cell. Longev. 2022, 2022, 3848084. [Google Scholar] [CrossRef] [PubMed]
  187. Vattem, D.; Shetty, K. Biological functionality of ellagic acid: A review. J. Food Biochem. 2005, 29, 234–266. [Google Scholar] [CrossRef]
  188. Gourineni, V.; Shay, N.F.; Chung, S.; Sandhu, A.K.; Gu, L. Muscadine grape (Vitis rotundifolia) and wine phytochemicals prevented obesity-associated metabolic complications in C57BL/6J mice. J. Agric. Food Chem. 2012, 60, 7674–7681. [Google Scholar] [CrossRef]
  189. Cao, K.; Xu, J.; Pu, W.; Dong, Z.; Sun, L.; Zang, W.; Gao, F.; Zhang, Y.; Feng, Z.; Liu, J. Punicalagin, an active component in pomegranate, ameliorates cardiac mitochondrial impairment in obese rats via AMPK activation. Sci. Rep. 2015, 5, 14014. [Google Scholar] [CrossRef] [PubMed]
  190. Kahn, B.B.; Alquier, T.; Carling, D.; Hardie, D.G. AMP-activated protein kinase: Ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005, 1, 15–25. [Google Scholar] [CrossRef]
  191. Kang, I.; Espín, J.C.; Carr, T.P.; Tomás-Barberán, F.A.; Chung, S. Raspberry seed flour attenuates high-sucrose diet-mediated hepatic stress and adipose tissue inflammation. J. Nutr. Biochem. 2016, 32, 64–72. [Google Scholar] [CrossRef] [PubMed]
  192. Slatnar, A.; Mikulic-Petkovsek, M.; Stampar, F.; Veberic, R.; Solar, A. Identification and quantification of phenolic compounds in kernels, oil and bagasse pellets of common walnut (Juglans regia L.). Food Res. Int. 2015, 67, 255–263. [Google Scholar] [CrossRef]
  193. Gómez-Caravaca, A.M.; Verardo, V.; Segura-Carretero, A.; Caboni, M.F.; Fernández-Gutiérrez, A. Development of a rapid method to determine phenolic and other polar compounds in walnut by capillary electrophoresis–electrospray ionization time-of-flight mass spectrometry. J. Chromatogr. A 2008, 1209, 238–245. [Google Scholar] [CrossRef]
  194. Regueiro, J.; Sánchez-González, C.; Vallverdú-Queralt, A.; Simal-Gándara, J.; Lamuela-Raventós, R.; Izquierdo-Pulido, M. Comprehensive identification of walnut polyphenols by liquid chromatography coupled to linear ion trap–Orbitrap mass spectrometry. Food Chem. 2014, 152, 340–348. [Google Scholar] [CrossRef]
  195. Ren, S.-m.; Zhang, Q.-z.; Chen, M.-l.; Jiang, M.; Zhou, Y.; Xu, X.-j.; Wang, D.-m.; Pan, Y.-n.; Liu, X.-q. Anti-NAFLD effect of defatted walnut powder extract in high fat diet-induced C57BL/6 mice by modulating the gut microbiota. J. Ethnopharmacol. 2021, 270, 113814. [Google Scholar] [CrossRef]
  196. Elseweidy, M.M.; Elesawy, A.E.; Sobh, M.S.; Elnagar, G.M. Ellagic acid ameliorates high fructose-induced hyperuricemia and non-alcoholic fatty liver in Wistar rats: Focusing on the role of C1q/tumor necrosis factor-related protein-3 and ATP citrate lyase. Life Sci. 2022, 305, 120751. [Google Scholar] [CrossRef]
  197. Mandal, A.K.; Mount, D.B. The molecular physiology of uric acid homeostasis. Annu. Rev. Physiol. 2015, 77, 323–345. [Google Scholar] [CrossRef]
  198. Lanaspa, M.A.; Sanchez-Lozada, L.G.; Choi, Y.-J.; Cicerchi, C.; Kanbay, M.; Roncal-Jimenez, C.A.; Ishimoto, T.; Li, N.; Marek, G.; Duranay, M. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: Potential role in fructose-dependent and-independent fatty liver. J. Biol. Chem. 2012, 287, 40732–40744. [Google Scholar] [CrossRef] [PubMed]
  199. Xie, D.; Zhao, H.; Lu, J.; He, F.; Liu, W.; Yu, W.; Wang, Q.; Hisatome, I.; Yamamoto, T.; Koyama, H. High uric acid induces liver fat accumulation via ROS/JNK/AP-1 signaling. Am. J. Physiol.-Endocrinol. Metab. 2021, 320, E1032–E1043. [Google Scholar] [CrossRef]
  200. Xu, K.; Zhao, X.; Fu, X.; Xu, K.; Li, Z.; Miao, L.; Li, Y.; Cai, Z.; Qiao, L.; Bao, J. Gender effect of hyperuricemia on the development of nonalcoholic fatty liver disease (NAFLD): A clinical analysis and mechanistic study. Biomed. Pharmacother. 2019, 117, 109158. [Google Scholar] [CrossRef]
  201. Yang, J.; Guo, Y.; Henning, S.M.; Chan, B.; Long, J.; Zhong, J.; Acin-Perez, R.; Petcherski, A.; Shirihai, O.; Heber, D. Ellagic Acid and Its Microbial Metabolite Urolithin A Alleviate Diet-Induced Insulin Resistance in Mice. Mol. Nutr. Food Res. 2020, 64, 2000091. [Google Scholar] [CrossRef] [PubMed]
  202. Jastroch, M.; Divakaruni, A.S.; Mookerjee, S.; Treberg, J.R.; Brand, M.D. Mitochondrial proton and electron leaks. Essays Biochem. 2010, 47, 53–67. [Google Scholar] [PubMed]
  203. B Chan, C.; Harper, M.-E. Uncoupling proteins: Role in insulin resistance and insulin insufficiency. Curr. Diabetes Rev. 2006, 2, 271–283. [Google Scholar] [CrossRef]
  204. Zhang, C.; Song, Y.; Yuan, M.; Chen, L.; Zhang, Q.; Hu, J.; Meng, Y.; Li, S.; Zheng, G.; Qiu, Z. Ellagitannins-Derived Intestinal Microbial Metabolite Urolithin A Ameliorates Fructose-Driven Hepatosteatosis by Suppressing Hepatic Lipid Metabolic Reprogramming and Inducing Lipophagy. J. Agric. Food Chem. 2023, 71, 3967–3980. [Google Scholar] [CrossRef]
  205. Fang, C.; Pan, J.; Qu, N.; Lei, Y.; Han, J.; Zhang, J.; Han, D. The AMPK pathway in fatty liver disease. Front. Physiol. 2022, 13, 970292. [Google Scholar] [CrossRef]
  206. Xu, J.; Tian, H.; Ji, Y.; Dong, L.; Liu, Y.; Wang, Y.; Gao, X.; Shi, H.; Li, H.; Yang, L. Urolithin C reveals anti-NAFLD potential via AMPK-ferroptosis axis and modulating gut microbiota. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2023, 396, 2687–2699. [Google Scholar] [CrossRef]
  207. Capelletti, M.M.; Manceau, H.; Puy, H.; Peoc’h, K. Ferroptosis in Liver Diseases: An Overview. Int. J. Mol. Sci. 2020, 21, 4908. [Google Scholar] [CrossRef]
  208. Mao, L.; Zhao, T.; Song, Y.; Lin, L.; Fan, X.; Cui, B.; Feng, H.; Wang, X.; Yu, Q.; Zhang, J.; et al. The emerging role of ferroptosis in non-cancer liver diseases: Hype or increasing hope? Cell Death Dis. 2020, 11, 518. [Google Scholar] [CrossRef]
  209. Abdulrahman, A.O.; Kuerban, A.; Alshehri, Z.A.; Abdulaal, W.H.; Khan, J.A.; Khan, M.I. Urolithins Attenuate Multiple Symptoms of Obesity in Rats Fed on a High-Fat Diet. Diabetes Metab. Syndr. Obes. 2020, 13, 3337–3348. [Google Scholar] [CrossRef]
  210. Buniatian, G. Stages of activation of hepatic stellate cells: Effects of ellagic acid, an inhibiter of liver fibrosis, on their differentiation in culture. Cell Prolif. 2003, 36, 307–319. [Google Scholar] [CrossRef]
  211. Lu, C.-C.; Yang, S.-H.; Hsia, S.-M.; Wu, C.-H.; Yen, G.-C. Inhibitory effects of Phyllanthus emblica L. on hepatic steatosis and liver fibrosis in vitro. J. Funct. Foods 2016, 20, 20–30. [Google Scholar] [CrossRef]
  212. Zhang, C.; Hu, J.; Sheng, L.; Yuan, M.; Wu, Y.; Chen, L.; Wang, G.; Qiu, Z. Ellagic acid ameliorates AKT-driven hepatic steatosis in mice by suppressing de novo lipogenesis via the AKT/SREBP-1/FASN pathway. Food Funct. 2019, 10, 3410–3420. [Google Scholar] [CrossRef]
  213. Ahmed, M.H.; Byrne, C.D. Modulation of sterol regulatory element binding proteins (SREBPs) as potential treatments for non-alcoholic fatty liver disease (NAFLD). Drug Discov. Today 2007, 12, 740–747. [Google Scholar] [CrossRef]
  214. O’Farrell, M.; Duke, G.; Crowley, R.; Buckley, D.; Martins, E.B.; Bhattacharya, D.; Friedman, S.L.; Kemble, G. FASN inhibition targets multiple drivers of NASH by reducing steatosis, inflammation and fibrosis in preclinical models. Sci. Rep. 2022, 12, 15661. [Google Scholar] [CrossRef]
  215. Cheon, S.Y.; Cho, K. Lipid metabolism, inflammation, and foam cell formation in health and metabolic disorders: Targeting mTORC1. J. Mol. Med. 2021, 99, 1497–1509. [Google Scholar] [CrossRef]
  216. Feng, J.; Qiu, S.; Zhou, S.; Tan, Y.; Bai, Y.; Cao, H.; Guo, J.; Su, Z. mTOR: A potential new target in nonalcoholic fatty liver disease. Int. J. Mol. Sci. 2022, 23, 9196. [Google Scholar] [CrossRef]
  217. Gheflati, A.; Mohammadi, M.; Ramezani-Jolfaie, N.; Heidari, Z.; Salehi-Abargouei, A.; Nadjarzadeh, A. Does pomegranate consumption affect weight and body composition? A systematic review and meta-analysis of randomized controlled clinical trials. Phytother. Res. 2019, 33, 1277–1288. [Google Scholar] [CrossRef]
  218. González-Sarrías, A.; Giménez-Bastida, J.A.; García-Conesa, M.T.; Gómez-Sánchez, M.B.; García-Talavera, N.V.; Gil-Izquierdo, A.; Sánchez-Álvarez, C.; Fontana-Compiano, L.O.; Morga-Egea, J.P.; Pastor-Quirante, F.A.; et al. Occurrence of urolithins, gut microbiota ellagic acid metabolites and proliferation markers expression response in the human prostate gland upon consumption of walnuts and pomegranate juice. Mol. Nutr. Food Res. 2010, 54, 311–322. [Google Scholar] [CrossRef]
  219. Falsaperla, M.; Morgia, G.; Tartarone, A.; Ardito, R.; Romano, G. Support Ellagic Acid Therapy in Patients with Hormone Refractory Prostate Cancer (HRPC) on Standard Chemotherapy Using Vinorelbine and Estramustine Phosphate. Eur. Urol. 2005, 47, 449–455. [Google Scholar] [CrossRef]
  220. Pantuck, A.J.; Leppert, J.T.; Zomorodian, N.; Aronson, W.; Hong, J.; Barnard, R.J.; Seeram, N.; Liker, H.; Wang, H.; Elashoff, R. Phase II study of pomegranate juice for men with rising prostate-specific antigen following surgery or radiation for prostate cancer. Clin. Cancer Res. 2006, 12, 4018–4026. [Google Scholar] [CrossRef]
  221. Ertam, I.; Mutlu, B.; Unal, I.; Alper, S.; Kivcak, B.; Ozer, O. Efficiency of ellagic acid and arbutin in melasma: A randomized, prospective, open-label study. J. Dermatol. 2008, 35, 570–574. [Google Scholar] [CrossRef]
  222. Kasai, K.; Yoshimura, M.; Koga, T.; Arii, M.; Kawasaki, S. Effects of Oral Administration of Ellagic Acid-Rich Pomegranate Extract on Ultraviolet-Induced Pigmentation in the Human Skin. J. Nutr. Sci. Vitaminol. 2006, 52, 383–388. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of ellagic acid and the formation of the gut microbial metabolite urolithins.
Figure 1. Chemical structures of ellagic acid and the formation of the gut microbial metabolite urolithins.
Antioxidants 13 00485 g001
Figure 2. Positive impact of Ellagic Acid demonstrated through an increase of the Nrf2 pathway and decreasing all other pathways and mechanisms to alleviate NAFLD/NASH.
Figure 2. Positive impact of Ellagic Acid demonstrated through an increase of the Nrf2 pathway and decreasing all other pathways and mechanisms to alleviate NAFLD/NASH.
Antioxidants 13 00485 g002
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

Senavirathna, T.; Shafaei, A.; Lareu, R.; Balmer, L. Unlocking the Therapeutic Potential of Ellagic Acid for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Antioxidants 2024, 13, 485. https://doi.org/10.3390/antiox13040485

AMA Style

Senavirathna T, Shafaei A, Lareu R, Balmer L. Unlocking the Therapeutic Potential of Ellagic Acid for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis. Antioxidants. 2024; 13(4):485. https://doi.org/10.3390/antiox13040485

Chicago/Turabian Style

Senavirathna, Tharani, Armaghan Shafaei, Ricky Lareu, and Lois Balmer. 2024. "Unlocking the Therapeutic Potential of Ellagic Acid for Non-Alcoholic Fatty Liver Disease and Non-Alcoholic Steatohepatitis" Antioxidants 13, no. 4: 485. https://doi.org/10.3390/antiox13040485

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