Hostname: page-component-8448b6f56d-tj2md Total loading time: 0 Render date: 2024-04-19T03:57:59.572Z Has data issue: false hasContentIssue false

Polyphenols and non-alcoholic fatty liver disease: impact and mechanisms

Published online by Cambridge University Press:  23 November 2015

I. Rodriguez-Ramiro*
Affiliation:
Department of Medicine, Norwich Medical School, University of East Anglia, Norwich, UK
D. Vauzour
Affiliation:
Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, UK
A. M. Minihane
Affiliation:
Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, UK
*
*Corresponding author: I. Rodriguez-Ramiro, email i.rodriguez-ramiro@uea.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Non-alcoholic fatty liver disease (NAFLD) is considered to be the hepatic component of the metabolic syndrome and its prevalence is rapidly increasing due to its strong association with insulin resistance and obesity. At present, given that NAFLD is highly prevalent and therapies are limited, much attention is focused on identifying effective dietary strategies for the prevention and treatment of the disease. Polyphenols are a group of plant bioactive compounds whose regular consumption have been associated with a reduction in the risk of a number of metabolic disorders associated with NAFLD. Here we review the emerging and relatively consistent evidence from cell culture and rodent studies showing that select polyphenols positively modulate a variety of contributors to the NAFLD phenotype, through diverse and complementary mechanisms of action. In particular, the reduction of de novo lipogenesis (via sterol regulatory element-binding protein 1c) and increased fatty acid β-oxidation, presumably involving AMP-activated protein kinase activation, will be discussed. The indirect antioxidant and anti-inflammatory properties of polyphenols which have been reported to contribute to the amelioration of NAFLD will also be addressed. In addition to a direct study of the liver, rodent studies have provided insight into the impact of polyphenols on adipose tissue function and whole body insulin sensitivity, which are likely to in part modulate their impact on NAFLD development. Finally an overview of the limited data from clinical trials will be given along with a discussion of the dose extrapolation from animal studies to human subjects.

Type
Conference on ‘Diet, gene regulation and metabolic disease’
Copyright
Copyright © The Authors 2015 

The term non-alcoholic fatty liver disease (NAFLD) refers to a condition defined by ectopic fat accumulation in the form of TAG in the liver, when it accounts for more than 5 % of the total organ weight. NAFLD encompasses a wide spectrum of liver damages, ranging from simple TAG accumulation in hepatocytes (steatosis) to non-alcoholic steatohepatitis (NASH), which is characterised by the additional presence of inflammation and tissue injury( Reference Byrne 1 , Reference Review, LaBrecque and Abbas 2 ). NASH can lead to fibrosis, which can progress to cirrhosis with a high risk of liver failure and hepatocellular carcinoma( Reference Schuppan, Gorrell and Klein 3 ). NAFLD is a major public health issue in industrialised countries( Reference Schuppan, Gorrell and Klein 3 ), with an estimated prevalence in the general population of 20–30 %( Reference Review, LaBrecque and Abbas 2 ). Most NAFLD patients are clinically asymptomatic with approximately 10–25 % progressing to NASH and 5–8 % of those will be susceptible to develop cirrhosis within 5 years. Furthermore, it has been reported that 12·8 % of patients with liver cirrhosis will develop hepatocellular carcinoma within 3 years( Reference Milic, Lulic and Stimac 4 ).

NAFLD is considered to be the hepatic component of the metabolic syndrome, which is characterised by insulin resistance, obesity (>90 % of NAFLD patients are overweight), hyperinsulinaemia, dyslipidaemia and hypertension( Reference Schuppan, Gorrell and Klein 3 , Reference Stojsavljevic, Gomercic Palcic and Virovic Jukic 5 , Reference Bhatia, Curzen and Calder 6 ). Besides it is a significant risk factor for CVD, which is the most prevalent clinical feature of NAFLD( Reference Bhatia, Curzen and Calder 6 ).

Although a persistent elevation of plasma transaminase enzymes can be used as an early indication of liver damage, the accurate diagnosis of NAFLD presence and severity is not possible by routine blood tests. For an accurate and sensitive diagnosis of NAFLD a liver biopsy accompanied by histological staining and NAFLD activity scoring is considered the gold standard, but its use in clinical practice is limited by its invasive nature( Reference Review, LaBrecque and Abbas 2 , Reference Berlanga, Guiu-Jurado and Porras 7 , Reference Kleiner, Brunt and Van Natta 8 ).

At present, NAFLD due to its high prevalence and pathological consequences, represents an important economic burden for European countries( Reference Blachier, Leleu and Peck-Radosavljevic 9 ). However, to date, there is no licensed medication or surgical procedure for NAFLD. Lifestyle strategies such as dietary and exercise regimens focused on weight reduction and insulin sensitisation have been the primary therapeutic approach( Reference Schuppan, Gorrell and Klein 3 ). Although these strategies have been shown to be efficacious in randomised controlled trials, at a population level, due to poor compliance, they have had a limited impact on NAFLD incidence and severity( Reference Schuppan, Gorrell and Klein 3 ). Therefore there is a great need to identify effective approaches for NAFLD management.

Polyphenols are found ubiquitously in plants and their regular consumption has been associated with a reduction in the risk of a number of metabolic diseases, including obesity, insulin resistance, hypertension and CVD( Reference Gu and Lambert 10 , Reference Rodriguez-Mateos, Vauzour and Krueger 11 ). New evidence supports the idea that a polyphenol-rich diet may have an important role in the prevention and treatment of NAFLD. The purpose of the present review is to consider the efficacy of polyphenols in NAFLD and to discuss the key molecular mechanisms which modulate their potential clinical benefits.

Non-alcoholic fatty liver disease pathophysiology

NAFLD has a complex pathophysiology, which is described by the two-hit hypothesis( Reference Berlanga, Guiu-Jurado and Porras 7 ). In this model, the first hit describes the accumulation of fatty acids (FA) and TAG in hepatocytes leading to steatosis, which results from multiple mechanisms such as: (a) increased hepatic delivery and uptake of FA associated with increased lipolysis in adipose tissue and/or increased intake of dietary fat; (b) decreased FA oxidation; (c) increased hepatic de novo lipogenesis; and (d) decreased hepatic lipid export via VLDL( Reference Berlanga, Guiu-Jurado and Porras 7 , Reference Tilg and Moschen 12 ). The inability to regulate lipid partitioning leads to the second hit, whereby an overwhelmed FA β-oxidation produces mitochondrial dysfunction which increases reactive oxygen species resulting in sustained oxidative stress and a depletion of the antioxidant defences( Reference Serviddio, Bellanti and Vendemiale 13 , Reference Gupte, Lyon and Hsueh 14 ). FA intermediates and a compromised oxidative status activates Kupffer cells producing inflammatory mediators, and dysregulated insulin action leading to the progression from benign steatosis to NASH( Reference Schuppan, Gorrell and Klein 3 , Reference Serviddio, Bellanti and Vendemiale 13 ). Finally, chronic inflammation and oxidative stress induce hepatocyte apoptosis and injury which activates stellate cells which are central to the progression to liver fibrosis( Reference Schuppan, Gorrell and Klein 3 , Reference Gupte, Lyon and Hsueh 14 ).

Polyphenols: chemical structures and sources

Phenolic compounds are secondary metabolites of plants which are present in high amounts in fruits, vegetables, cereals and beverages such as red wine, tea or coffee. More than 8000 structures have been identified ranging from compounds with at least one aromatic ring with one or two hydroxyl groups, to polymers of up to fifty units with multiple hydroxyl groups. Generally, all phenolic compounds are commonly referred to as polyphenols, despite a group of them having only one aromatic ring. Polyphenols are divided into two main categories, namely flavonoids and non-flavonoids, based on the number of phenol rings and the way in which these rings interact( Reference Del Rio, Rodriguez-Mateos and Spencer 15 ).

Flavonoids have a common basic structure of fifteen carbons (C6–C3–C6) with two aromatic carbon rings (A and B rings) connected by a three-carbon bridge (C ring). Flavonoids may be sub-classified according to the degree of oxidation of the C-ring, the hydroxylation pattern of the ring structure and the substitution of the three-position into: (a) flavonols (e.g. quercetin and kaempferol) whose sources include onions and broccoli, (b) flavones (e.g. luteolin, apigenin) found in celery and parsley, (c) isoflavones (e.g. genistein and daidzein) found in leguminous plants and in particular soyabeans and soya products, (d) flavanones (e.g. naringerin and hesperitin) abundant in citrus fruits, wine and herbs such as oregano, (e) anthocyanidins (e.g. cyanidin and peonidin) found in berry fruits and red wine, and (f) flavanols (e.g. (+)-catechin, (−)-epicatechin, epigallocatechin) abundant in cocoa and green tea( Reference Rodriguez-Mateos, Vauzour and Krueger 11 , Reference Del Rio, Rodriguez-Mateos and Spencer 15 ) (Fig. 1).

Fig. 1. Polyphenol structures.

Non-flavonoids may be sub-classified into phenolic acids and stilbenes. Phenolic acid includes hydroxybenzoic acids (C6–C1) and hydroxycinnamic acids (C6–C3). Hydroxybenzoic acids (e.g. gallic acid) are found in pomegranate and raspberries. Hydroxycinnamic acids (e.g. caffeic acid) can be found in coffee beans and blueberries. Stilbenes have a C6–C2–C6 structure. Resveratrol which is the main stilbene, can be found as cis or trans isomers as well as conjugated derivatives in grapes and red wine( Reference Rodriguez-Mateos, Vauzour and Krueger 11 , Reference Del Rio, Rodriguez-Mateos and Spencer 15 ) (Fig. 1).

Polyphenols have been identified as powerful antioxidants in vitro ( Reference Williams, Spencer and Rice-Evans 16 ). However, given their extensive metabolism and relatively low tissue concentrations, their in vivo preventative properties are considered largely independent of conventional antioxidant activities( Reference Williams, Spencer and Rice-Evans 16 ). The ability of polyphenols to exert antioxidant properties in vivo depends on the extent of their phases 1 and 2 biotransformation and conjugation products during absorption in the gastrointestinal tract and post-absorption primarily in the liver. Although a full overview of polyphenols metabolism and its regulation is beyond the scope of the current review (see Rodriguez-Mateos et al. ( Reference Rodriguez-Mateos, Vauzour and Krueger 11 ) for an extensive review), knowledge about their bio-kinetics (the composite of their distribution, biotransformation and elimination) alluded to throughout, is essential to understand the bioactivity of polyphenols in vivo ( Reference Rodriguez-Mateos, Vauzour and Krueger 11 ).

In vitro studies

Cell culture studies constitute a useful tool to elucidate the molecular mechanisms of action of polyphenols in the prevention of steatosis. Primary cultures of human hepatocytes are the optimal cell culture model for studying determinants of NAFLD. However their widespread use is limited by logistical factors such as liver samples availability. The main alternative model is the human hepatocyte-derived cell line, HepG2.

Palmitic (16 : 0) and oleic (18 : 1n-9) acids are the most abundant FA in the liver of both normal subjects and NAFLD patients( Reference Gomez-Lechon, Donato and Martinez-Romero 17 ) and have been used (generally in a bovine serum complex) to induce lipid accumulation in HepG2 and reproduce the key cellular features of human NAFLD( Reference Gomez-Lechon, Donato and Martinez-Romero 17 Reference Shimada, Tokuhara and Tsubata 19 ). In addition, steatosis in HepG2 cells has been induced by high concentrations of glucose (25–30 mm)( Reference Pil Hwang, Gyun Kim and Choi 20 , Reference Shang, Chen and Xiao 21 ) which through a multistep process, including glycolysis and the Krebs Cycle generates acetyl-CoA, a key substrate for de novo lipogenesis( Reference Berlanga, Guiu-Jurado and Porras 7 ).

Pure polyphenol compounds and polyphenol-rich extracts have been tested in both these in vitro models of steatosis (Table 1). Most studies are concordant with the fact that a range of polyphenols reduce hepatocellular TAG accumulation induced by FA( Reference Vidyashankar, Sandeep Varma and Patki 18 , Reference Shimada, Tokuhara and Tsubata 19 , Reference Liu, Ma and Wang 22 Reference Wu, Lin and Wang 24 ) or by high glucose concentrations( Reference Pil Hwang, Gyun Kim and Choi 20 , Reference Shang, Chen and Xiao 21 , Reference Guo, Li and Ling 25 ) with a range of reported mechanisms, including an inhibition of lipogenesis and a promotion of FA catabolism (Fig. 2).

Fig. 2. (Colour online) Possible mechanisms underlying the effect of polyphenols in non-alcoholic fatty liver disease (NAFLD). Polyphenols may prevent cellular damage in hepatocytes associated with NAFLD through different mechanism of action including: (a) reducing de novo lipogenesis through sterol regulatory element-binding protein 1c (SREBP-1c) down-regulation, (b) increasing β-fatty acid (FA) oxidation by PPARα up-regulation, (c) improving insulin sensitivity (d) reducing oxidative stress through increasing the antioxidant defence levels via nuclear factor-erythroid 2-related factor 2 (Nrf2), (e) attenuating the inflammatory pathways. Presumably SREBP-1c down-regulation and PPARα up-regulation are modulated by AMPK activation (by phosphorylation). TNFR, TNFα receptor; IL6-R, IL-6 receptor; IR, insulin receptor; CD36, cluster of differentiation 36/FA translocase; p-AMPK, phosphorylated AMP-activated protein kinase α; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; SCD, stearoyl-CoA desaturase; GPAT, glycerol-3-phosphate acyltransferase; CPT-1, carnitine palmitoyl transferase 1; ACO, acyl-CoA oxidase; PGC-1, PGC1α, PPARγ coactivator-1α; JNK, c-Jun N-terminal kinase; PKC, protein kinase C; mTOR, mammalian target of rapamycin.

Table 1. Cell culture studies investigating the impact of polyphenols on non-alcoholic fatty liver disease

Arrow indicates an increase (↑) or decrease (↓) in the levels of gene expression, protein concentrations or activity.

p-AMPKα, phosphorylated AMP-activated protein kinase α; ACC, acetyl-CoA carboxylase; CPT-1, carnitine palmitoyl transferase 1; FAS, fatty acid synthase; ROS, reactive oxygen species; SREBP-1c, sterol regulatory element-binding protein 1c; p-ACC, phosphorylated ACC; SIRT-1, sirtuin-1; GSH, reduced glutathione; GSSG, oxidised glutathione; TNFα, tumor necrosis factor α; CAT, catalase; SOD, superoxide dismutase; GPx, glutathione peroxidase; ALT, alanine aminotransferase; HGMCR, 3-hydroxy-3-methylglutaryl coenzyme A reductase; GPAT, glycerol-3-phosphate acyltransferase; mtGPAT, mitochondrial GPAT; PKC, protein kinase C; p-JNK, phosphorylated, c-Jun N-terminal kinase; MDA, malondialdehyde; HFD, high-fat diet.

Sterol regulatory element-binding protein 1c (SREBP-1c) is the most important transcription factor regulating genes involved in FA synthesis and TAG metabolism in the liver( Reference Jump, Tripathy and Depner 26 ). A number of in vitro studies with polyphenols have shown a down-regulation of SREBP-1c and its main targets in lipogenesis( Reference Pil Hwang, Gyun Kim and Choi 20 , Reference Liu, Ma and Wang 22 , Reference Wu, Lin and Wang 24 ). In particular, Liu et al.( Reference Liu, Ma and Wang 22 ) reported that luteolin induced a reduction of palmitate-stimulated lipid accumulation in HepG2 cells associated with decreased SREBP-1c and FA synthase gene expression and an attenuation of the activity of acetyl-CoA carboxylase. Acetyl-CoA carboxylase and FA synthase play an essential role in de novo lipogenesis converting the acetyl-CoA into palmitate that subsequently is esterified into TAG in the liver. Similar reduced expression of SREBP-1c and FA synthase were reported using a chlorogenic acid derivative (3-caffeoyl, 4-dihydrocaffeoylquinic acid) and rutin (quercetin-3-O-rutinoside) in a high glucose-stimulated and oleic-stimulated lipid accumulation HepG2 cell model, respectively( Reference Pil Hwang, Gyun Kim and Choi 20 , Reference Wu, Lin and Wang 24 ). In addition, treatment with 3-caffeoyl, 4-dihydrocaffeoylquinic acid, luteolin and rutin induced an activation (by phosphorylation) of AMP-activated protein kinase (AMPK), a well-known inhibitor of SREBP-1c and hence of lipogenesis( Reference Pil Hwang, Gyun Kim and Choi 20 , Reference Liu, Ma and Wang 22 , Reference Wu, Lin and Wang 24 ). Sirtuin 1 (SIRT-1) activation by polyphenols represents a downstream regulator of AMPK( Reference Canto and Auwerx 27 ). Pil et al. ( Reference Pil Hwang, Gyun Kim and Choi 20 ) found that 3-caffeoyl, 4-dihydrocaffeoylquinic acid treatment increased SIRT-1 activity, suggesting that SIRT-1 may be involved in the AMPK-dependent reduction in SREBP-1c and FA synthase expression induced by polyphenols. Cyanidin-3-O-β-glucoside also attenuated de novo lipogenesis through an alternative pathway, increasing protein kinase C ζ activity and suppressing mitochondrial glycerol-3-phosphate acyltransferase 1 activation, the rate limiting enzyme which controls the first step of TAG synthesis from palmitate( Reference Guo, Li and Ling 25 ).

In the liver, PPARα plays a pivotal role in FA metabolism by up-regulating the expression of numerous genes involved in FA oxidation as well as other processes which regulate cellular FA status such as receptor mediated FA uptake and lipoprotein assembly and secretion( Reference Contreras, Torres and Tovar 28 ). As a consequence, activation of PPARα is associated with decreased hepatic fat storage( Reference Berlanga, Guiu-Jurado and Porras 7 ). Oxidation of FA occurs within the mitochondria, peroxisomes and the endoplasmic reticulum and is regulated mainly through key rate limiting enzymes such as carnitine palmitoyl transferase 1 (CPT-1) and acyl-CoA oxidase. In the outer membrane of mitochondria, (CPT-1) mediates the transfer of FA from the cytosol into the mitochondria prior to β-oxidation and acyl-CoA oxidase catalyses the first rate-limiting step in peroxisomal β-oxidation( Reference Berlanga, Guiu-Jurado and Porras 7 , Reference Serviddio, Bellanti and Vendemiale 13 ). Procyanidin B1 (an epicatechin-(4β→8)-catechin dimer) suppressed palmitic-stimulated lipid accumulation in HepG2 cells through an up-regulation of the acyl-CoA oxidase and (CPT-1) mRNA expression( Reference Shimada, Tokuhara and Tsubata 19 ). In addition to inhibiting lipogenesis, luteolin induced (CPT-1) gene expression in HepG2 challenged with palmitate( Reference Liu, Ma and Wang 22 ). Furthermore, rutin increased PPARα protein levels which was associated with a reduction in the lipid load in HepG2 cells( Reference Wu, Lin and Wang 24 ).

It is well known that a number of polyphenols can indirectly act as antioxidants by inducing phase II antioxidant defences enzymes( Reference Masella, Di Benedetto and Vari 29 Reference Rodriguez-Ramiro, Martin and Ramos 31 ). There is evidence suggesting that the antioxidant response can alleviate the cellular damage induced by oxidative stress during the progression of NAFLD( Reference Gupte, Lyon and Hsueh 14 ). Accordingly, Vidyashankar et al.( Reference Vidyashankar, Sandeep Varma and Patki 18 ) reported that quercetin induced an increase in the activity of antioxidant cellular defences, such as catalase, glutathione peroxidase and superoxide dismutase and an increase of reduced glutathione levels. Likewise, rutin attenuated the cellular oxidative stress induced by oleic acid through raised superoxide dismutase, glutathione peroxidase and catalase protein levels which was associated with an increase in PPARα protein levels( Reference Wu, Lin and Wang 24 ). A sustained oxidative stress can induce hepatocyte apoptosis and accentuate the transition from simple steatosis to NASH. Jiang et al., showed that cyanidin-3-O-β-glucoside reduced oxidative stress and the apoptotic pathway activation induced by hyperglycaemia, preventing mitochondrial dysfunction through modulation of phosphatidylinositol-3-kinase/protein kinase B and c-Jun N-terminal kinase (JNK) -signalling pathways( Reference Jiang, Tang and Zhang 32 ).

Animal in vivo studies

Animal models of NAFLD can be divided in three major categories: those caused by a genetic mutation, by a dietary or pharmacological manipulation, or a combination of both models( Reference Takahashi, Soejima and Fukusato 33 ). The choice of model results in variability in the characteristics and severity of the NAFLD phenotype and its aetiological basis, with careful selection needed in order to address the specific research question in a meaningful way. For example, two of the most widely used dietary models of NASH, the high fat diet (HFD) and the methionine and choline deficiency (MCD) models display important differences in their metabolic characteristics. Although both present significant steatosis, mice fed a HFD develop obesity and insulin resistance which are characteristic of NAFLD and NASH in human subjects. Conversely mice fed a MCD exhibit atypical (for human subjects) weight loss and low serum insulin and leptin levels. However, the MCD model produces a more pathological form of NAFLD characterised by severe inflammation, oxidative stress, mitochondrial dysfunction, apoptosis and fibrogenesis, features which are only induced to a limited extent when using the HFD model( Reference Takahashi, Soejima and Fukusato 33 ). For evaluation of the efficacy of dietary approaches in NAFLD, the HFD may be chosen when evaluating the ability to prevent NAFLD development or for ameliorating steatosis, whereas the MCD model may be more appropriate to assess the therapeutic potential to reverse NASH associated liver injury.

Several studies have revealed that different subclasses of polyphenols ameliorate the severity and metabolic consequences of NAFLD in animal models. In general, liver biopsies (using haematoxylin/eosin staining) accompanied by semi-quantitative NAFLD activity scoring have shown that pure polyphenols or polyphenolic extracts reduced liver TAG accumulation( Reference Ueno, Torimura and Nakamura 34 Reference Joven, Espinel and Rull 38 ). However, the underlying molecular mechanisms associated with reduced steatosis are variable and dependant on the choice of animal model and the dose of phenolic compound of interest (Table 2 and Fig. 2).

Table 2. Rodent studies evaluating the impact of polyphenols on non-alcoholic fatty liver disease

Arrow indicates an increase (↑) or decrease (↓) in the levels of expression or activity.

EGCG, epigallocatequin-3-gallate; ALT, alanine aminotransferase; SREBP-1c, sterol regulatory element-binding protein 1c; IRS-1, insulin receptor substrate-1; p-IRS-1, phosphorylated IRS-1; p-AKT, phosphorylated AKT; p-IKKβ, phosphorylated inhibitor of κB kinase β subunit; HFD, high fat diet; AMPK, adenosine monophosphate-activated protein kinase; ACC, acetyl-CoA carboxylase; LXRα, liver X receptor α; FAS, fatty acid synthase; CPT-1, carnitine palmitoyl transferase; TBARS, thiobarbituric acid reactive substances; p-JNK, phosphorylated, c-Jun N-terminal kinase; ; IκB, inhibitor of κB; AST, aspartate aminotransferase; CAT, catalase; αSMA, α-smooth muscle actin-positive cells; TLR, toll-like receptor; p-AMPK, phosphorylated adenosine monophosphate-activated protein kinase; NOS, nitric oxide synthase, SOD, superoxide dismutase; GPx, glutathione peroxidase; ALP, alkaline phosphatase; UCP2, uncoupling protein 2; PGC1α, PPARγ coactivator-1α; GSH, reduced glutathione ; GSSG, oxidised glutathione; ACO, acyl-Coenzyme A oxidase; SIRT-1, sirtuin-1; p-ACC, phosphorylated ACC, ChREBP, carbohydrate-responsive element-binding protein; SCD-1, stearoyl-CoA desaturase; ACAT, acyl-CoA:colesterol acyltransferase; ME, malic enzyme; G6PD, glucose-6-phosphate dehydrogenase; MDA, malondialdehyde; FRAP, ferric reducing antioxidant power; miR, microRNA; Q3G, quercetin 3-O-β-D-glucuronide; GPAT1, glycerol-3-phosphate acyltransferase 1.

Adipokine amelioration

NAFLD has been correlated with visceral adiposity and dysregulation of a variety of adipokines( Reference Hui, Xu and Bo Yang 39 ). Increased serum leptin levels are found in NAFLD patients and are correlated with the severity of hepatic steatosis( Reference Huang, Fan and Zhang 40 ). Adiponectin has been recently reported to hamper the excess lipid storage in the liver and decreased levels of this adipokine are observed in NASH patients( Reference Buechler, Wanninger and Neumeier 41 ). In HFD-fed mice, dietary intake of the isoflavone genistein has been shown to reduce hepatic steatosis and adiposity. This ‘anti-adiposity’ effect has been associated with a modulation of adipokines gene expression, reducing leptin levels and increasing adiponectin levels in the adipose tissue( Reference Kim, Kang and Lee 42 ). Likewise, in the HFD-fed mice model, polyphenol-rich grape extract supplementation ameliorated abnormal plasma leptin and adiponectin levels which were associated with a reduction in NEFA( Reference Park, Jung and Lee 43 ). Collectively these results suggest that polyphenols could partially prevent the hepatic steatosis associated with obesity through improved regulation of adipokines.

Improvement of insulin sensitivity and de novo lipogenesis reduction

Postprandial insulin secretion promotes hepatic glucose uptake, and glycogen synthesis inhibits gluconeogenesis and stimulates de novo lipogenesis through SREBP-1c activation( Reference Serviddio, Bellanti and Vendemiale 13 ). In obese-hyperinsulinaemic mice, insulin signalling fails to decrease gluconeogenesis but still stimulates lipogenesis through SREBP-1c up-regulation, producing liver hypertriglyceridaemia and hyperglycaemia( Reference Brown and Goldstein 44 ). Using different NAFLD rodent models, resveratrol, genistein and an anthocyanin rich Hibiscus sabdariffa L. extract (HSE) have been shown to reduce insulin levels( Reference Shang, Chen and Xiao 21 , Reference Joven, Espinel and Rull 38 , Reference Huang, Qiao and Dong 45 ) along with reducing de novo lipogenic gene and protein expression and their master regulator SREBP-1c( Reference Joven, Espinel and Rull 38 , Reference Kim, Kang and Lee 42 , Reference Park, Jung and Lee 43 , Reference Huang, Qiao and Dong 45 Reference Tsuruta, Nagao and Kai 47 ). In addition, in nSREBP-1c transgenic C57/BL6 male mice, which show severe insulin resistance and develop NASH, an epigallocatechin-3-gallate supplement improved insulin sensitivity and promoted the functional recovery of insulin receptor substrate-1( Reference Ueno, Torimura and Nakamura 34 ).

Enhancement of β-fatty acid oxidation

An imbalance between lipogenesis and FA oxidation is central to the development and progression of steatosis/NASH. In this regard, an increase in the liver SREBP-1c:PPARα ratio, due to an up-regulation of SREBP-1c and/or down-regulation of PPARα, has been proposed to favour the development of steatosis in obese patients with NAFLD( Reference Pettinelli, Del Pozo and Araya 48 ). In mice fed an HFD, anthocyanin-rich juice supplementation stimulated PPARα up-regulation in parallel with a down-regulation of de novo lipogenic genes expression in the liver( Reference Salamone, Li Volti and Titta 49 ). Supplementation with isoflavones reduced liver steatosis by up-regulating genes involved in FA β-oxidation and down-regulating genes associated with lipogenesis in the adipose tissue( Reference Kim, Kang and Lee 42 ). Vitaglione et al.( Reference Vitaglione, Morisco and Mazzone 50 ) have also reported an up-regulation of PPARα gene expression and a higher rate of β-oxidation in the liver of rats with NASH supplemented with coffee polyphenols extract as a mechanism to reduce fat deposition in the liver. In addition, resveratrol supplementation in rats fed a high fat–high sucrose diet activated PPARγ co-activator 1α, a co-factor of PPARα in the induction of mitochondrial oxidative metabolism, associated with an increase in β-FA oxidation( Reference Alberdi, Rodriguez and Macarulla 51 )

Adenosine monophosphate-activated protein kinase as a key regulator in non-alcoholic fatty liver disease prevention

There is evidence that activation of AMPK is a central target for the effects of polyphenols in metabolic disorders related to NAFLD( Reference Um, Park and Kang 52 ). Consistent with this assumption, Beltran-Debón et al. ( Reference Joven, Espinel and Rull 38 , Reference Beltran-Debon, Rull and Rodriguez-Sanabria 53 ) have demonstrated that HSE and Rooibos extracts can prevent steatosis through AMPK activation in LDL receptor deficient mice (LDLr −/−) fed a high fat–high cholesterol diet. Similarly, other studies have reported that the preventative effect of resveratrol on liver fat accumulation, through up-regulation of FA oxidation and down-regulation of lipogenesis, was at least in part mediated by the activation of the AMPK/SIRT-1 axis( Reference Shang, Chen and Xiao 21 , Reference Alberdi, Rodriguez and Macarulla 51 ). It has also been reported that AMPK in the liver enhances the ratio between β-oxidation and lipogenesis, via SREBP-1c down-regulation( Reference Viollet, Guigas and Leclerc 54 ) and a promotion of mitochondrial content and function( Reference Canto and Auwerx 27 ). Furthermore, AMPK stimulates β-FA oxidation indirectly through inhibition of acetyl-CoA carboxylase which synthesises malonyl-CoA from acetyl-CoA( Reference Hardie and Pan 55 ). Malonyl-CoA has been described as an allosteric inhibitor of carnitine palmitoyl transferase 1( Reference Mills, Foster and McGarry 56 ). Therefore, acetyl-CoA carboxylase inactivation by AMPK reduces TAG synthesis but also enhances the FA influx to the mitochondria for β-FA oxidation( Reference Hardie and Pan 55 ). In consequence, the activation of AMPK by polyphenols has emerged as an important target in the prevention of NAFLD.

Antioxidant defences mechanisms prevent non-alcoholic fatty liver disease progression

NAFLD is characterised by oxidative stress and a redox imbalance generated in part as a consequence of insulin resistance and an accumulation of FA in hepatocytes( Reference Schuppan, Gorrell and Klein 3 , Reference Serviddio, Bellanti and Vendemiale 13 ). Elevated free radicals, lipid peroxidation and reduced antioxidants have been observed in NAFLD patients and animals models( Reference Serviddio, Bellanti and Vendemiale 13 ). Nuclear factor-erythroid 2-related factor 2 (Nrf2) is the main transcription factor which maintains cellular redox status through downstream modulation of antioxidant defences genes( Reference Gupte, Lyon and Hsueh 14 ). It has been recently reported that Nrf2 knockout mice (Nrf2−/−) fed a HFD developed a more severe steatosis and inflammation than wild-type Nrf2 mice( Reference Meakin, Chowdhry and Sharma 57 , Reference Cui, Wang and Li 58 ) which indicates the hepato-protective role of Nrf2. It is widely accepted that numerous polyphenols can activate Nrf2 which in turn, induce a variety of antioxidant defence enzymes which would result in reduced oxidative stress( Reference Masella, Di Benedetto and Vari 29 , Reference Rodriguez-Ramiro, Ramos and Bravo 30 ). Consistent with this statement, supplementation with quercetin, resveratrol and genistein have been reported to reduce lipid peroxidation in both the liver( Reference Ji, Yang and Hao 35 Reference Gomez-Zorita, Fernandez-Quintela and Macarulla 37 , Reference Li, Hai and Li 59 , Reference Bujanda, Hijona and Larzabal 60 ) and serum( Reference Ji, Yang and Hao 35 ) of NAFLD animals. Gomez-Zorita et al. ( Reference Gomez-Zorita, Fernandez-Quintela and Macarulla 37 ) also reported a raised reduced- glutathione:oxidised glutathione ratio level and Bujanda et al. ( Reference Bujanda, Hijona and Larzabal 60 ) an increase in the catalase, superoxide dismutase and glutathione peroxidase enzymatic activities in the liver of the NAFLD animals fed with resveratrol.

Anti-inflammatory effect preventing non-alcoholic fatty liver disease onset and progression

Inflammation is one of the main hallmarks of the progression from steatosis to NASH. It has been proposed that obesity promotes a systemic chronic low-grade inflammation which contributes to the development of metabolic disorders such as NAFLD( Reference Milic, Lulic and Stimac 4 ). TNFα and IL-6 are two of the main pro-inflammatory cytokines involved in the onset and progression of NAFLD which are secreted initially in the adipose tissue and later in the liver by Kupffer cells( Reference Stojsavljevic, Gomercic Palcic and Virovic Jukic 5 , Reference Hui, Xu and Bo Yang 39 ). It has been described that the interaction of TNFα with its receptor inhibits insulin receptors and activates NF-κB transcription factor and JNK pathways( Reference Tilg and Moschen 12 ). In addition increased hepatic and circulating TNFα and IL-6 levels have been observed in patients with NAFLD( Reference Berlanga, Guiu-Jurado and Porras 7 , Reference Tilg and Moschen 12 ). Recently, it has been proposed that a HFD can alter gut microbiota speciation and metabolism which e.g. via alterations in lipopolysaccharide production, can influence not only gastrointestinal, but also systemic inflammation( Reference Bleau, Karelis and St-Pierre 61 ). In rodent models supplementation with different polyphenols reduced the inflammatory profile in the serum/liver induced by HFD or MCD contributing to the amelioration of fatty liver dysfunction( Reference Ji, Yang and Hao 35 , Reference Andrade, Paraiso and de Oliveira 46 , Reference Vitaglione, Morisco and Mazzone 50 , Reference Li, Hai and Li 59 , Reference Marcolin, San-Miguel and Vallejo 62 ). In particular, studies using genistein, quercetin and resveratrol suggested that this anti-inflammatory effect was achieved through the repression of NF-κB translocation or gene expression( Reference Ji, Yang and Hao 35 , Reference Andrade, Paraiso and de Oliveira 46 , Reference Marcolin, San-Miguel and Vallejo 62 ) as well as a diminution in the JNK phosphorylation protein levels( Reference Ji, Yang and Hao 35 , Reference Marcolin, San-Miguel and Vallejo 62 ). Adiponectin is also involved in the anti-inflammatory response( Reference Berlanga, Guiu-Jurado and Porras 7 , Reference Hui, Xu and Bo Yang 39 ). The enhanced adiponectin secretion and gene expression induced by polyphenol-rich grape extract( Reference Park, Jung and Lee 43 ) and genistein( Reference Kim, Kang and Lee 42 ) (see earlier) may also contribute to reduced hepatic inflammation and ultimately the progression of NAFLD.

Clinical trials

To the best of our knowledge, only five human randomised controlled trials (all with a double-blinded placebo-controlled design) focused on polyphenols and NAFLD, have been published to date (Table 3). Three were undertaken with 500 and 600 mg resveratrol for 12 weeks( Reference Faghihzadeh, Adibi and Rafiei 63 , Reference Chen, Zhao and Ran 64 ) or 3000 mg for 8 weeks( Reference Chachay, Macdonald and Martin 65 ). The other two studies were carried out using an HSE (about 150 mg polyphenols)( Reference Chang, Peng and Yeh 66 ) or a bayberry juice (500 ml equivalent to 1350 mg polyphenols)( Reference Guo, Zhong and Liu 67 ) for 12 and 4 weeks, respectively. Four out of the five studies have reported a significant impact of intervention on select characteristics of NAFLD. Chang et al. reported that anthropometric measures (body weight, BMI and waist:hip ratio) were significantly lower (1·4, 1·33 and 1·09 %, respectively) following intervention with HSE( Reference Chang, Peng and Yeh 66 ) but no changes were observed with bayberry juice( Reference Guo, Zhong and Liu 67 ). For the two clinical trials using a similar dose of resveratrol (500 and 600 mg) only one observed a reduction in anthropometric measurements. This apparent discrepancy is likely due to the fact that in one of the trials resveratrol intervention was accompanied by a change in lifestyle with patients advised to follow physical activity guidelines( Reference Faghihzadeh, Adibi and Rafiei 63 ). With regard to hepatic function, two of the resveratrol interventions reduced the serum alanine transaminase concentrations by 15 %( Reference Faghihzadeh, Adibi and Rafiei 63 , Reference Chen, Zhao and Ran 64 ) although no reduction was detected in the studies with other polyphenol extracts( Reference Chang, Peng and Yeh 66 , Reference Guo, Zhong and Liu 67 ). In addition, one of the interventions with resveratrol and the HSE showed a reduction in serum total- and LDL-cholesterol( Reference Chen, Zhao and Ran 64 ) and NEFA( Reference Chang, Peng and Yeh 66 ). A significant reduction in the homeostasis model assessment insulin resistance index associated with lower serum glucose levels following resveratrol supplementation was also reported( Reference Chen, Zhao and Ran 64 ). The clinical trials using the bayberry juice and resveratrol reported anti-inflammatory effects, with a reduction in serum cytokines (TNFα, IL-6 and IL-8)( Reference Faghihzadeh, Adibi and Rafiei 63 , Reference Chen, Zhao and Ran 64 , Reference Guo, Zhong and Liu 67 ) and increased serum adiponectin levels( Reference Chen, Zhao and Ran 64 ). In addition, one of the interventions with resveratrol reported a reduction in NF-κB activity in the peripheral blood mononuclear cells( Reference Faghihzadeh, Adibi and Rafiei 63 ).

Table 3. Clinical trials carried out with polyphenols in non-alcoholic fatty liver disease (NAFLD) subjects

Arrow indicates increase (↑) and decrease (↓) in the levels of expression or activity.

HOMA-IR, homeostasis model assessment insulin resistance index; TNFα, tumour necrosis factor alpha; CRP, C-reactive protein; AST: aspartate aminotransferase; ALT, alanine aminotransferase

None of the clinical trials conducted liver biopsies and therefore had histological data on the severity of NAFLD. Instead non-invasive approaches such as semi-quantitative liver ultrasound examinations were carried out. Employing this approach, Chang et al. reported that HSE supplementation significantly reduced (by about 15 %) the liver damage score( Reference Chang, Peng and Yeh 66 ) and among the clinical trials with resveratrol, only the one accompanied by a change in lifestyle observed a significant reduction in steatosis( Reference Faghihzadeh, Adibi and Rafiei 63 , Reference Chen, Zhao and Ran 64 ). Finally, the non-beneficial effect of resveratrol observed at the higher supplementation dose( Reference Chachay, Macdonald and Martin 65 ) is likely due to a hormesis phenomenon, characterised by a low-dose stimulation and inhibition and a potentially detrimental effect at high-dose, which has been described for a number of bioactive compounds including resveratrol( Reference Calabrese, Mattson and Calabrese 68 ).

Doses of polyphenols: from animals studies to clinical trials

As discussed earlier, animal studies have been widely employed to assess the effects of a variety of polyphenols in NAFLD. However, the majority of these studies have used supra-physiological doses of compounds, with little consideration given to human equivalent doses( Reference Reagan-Shaw, Nihal and Ahmad 69 ). Taking resveratrol as an example, most of the pre-clinical studies in rats have employed doses ranging from 10 to 100 mg/kg body weight. Following allometric scaling calculations( Reference Reagan-Shaw, Nihal and Ahmad 69 ), such doses would equate to 97 and 970 mg resveratrol for a 60 kg person, although an estimated consumption in human subjects is only about 0·93 mg/d( Reference Zamora-Ros, Andres-Lacueva and Lamuela-Raventos 70 ). Therefore from a dose perspective the majority of the rodent scientific literature provides little insight into the likely benefits of dietary sourced resveratrol in human NAFLD, although such higher doses may be achievable through the consumption of resveratrol rich supplements.

However, the estimated intake of total polyphenols in Western populations is about 1–2 g/d with other polyphenols occurring in much higher amounts in the diet than resveratrol( Reference Rothwell, Perez-Jimenez and Neveu 71 ), with most plant sources consisting of a combination of different compounds which collectively may have a much greater impact on liver health relative to the effect of each one in isolation. Thus, more studies assessing possible additive and synergistic effects of polyphenol combinations commonly found in the diet are needed.

Conclusion

NAFLD is the major cause of chronic liver disease in Western countries and currently about 2–5 % of the population have NASH which is predicted to double by 2050( Reference Takahashi and Fukusato 72 , Reference Wree, Broderick and Canbay 73 ). As NAFLD is essentially a condition of overnutrition, and as there is a current lack of effective therapies, there is a great need to identify dietary approaches for NAFLD prevention and treatment. Taken together, the current cell and animal evidence suggests that a number of polyphenols could prevent steatosis and its progression to NASH. The mechanisms underlying such observations are likely to include improved adipokine regulation and insulin sensitivity, a decline in de novo lipogenesis (via SREBP-1c) and an increase in FA β-oxidation activity which would reduce the lipid load in the liver. Recent insights have proposed that the activation of the AMPK/SIRT-1 axis is the common trigger for the regulation of all these molecular processes. However, more experiments are required to verify this hypothesis. In addition, the indirect antioxidant and anti-inflammatory effects exerted by polyphenols are also likely to make a significant contribution to the amelioration of NAFLD. But to date results from clinical studies are limited and often shown a subtle effect in comparison with animal models. Further research in rodents and human subjects using dietary achievable doses of individual polyphenols or select combinations are needed.

Financial Support

This review was not supported by any funding agency in the public, commercial or not-for-profit sectors. A. M. M.'s ongoing research in the area of polyphenols and health is funded as part of a BBSRC ISP Grant (BB/J004545/1). I. R. R. is currently funded by a BBSRC grant (BB/L025396/1).

Conflict of Interest

None.

Authorship

I. R. R. wrote the manuscript and D. V. and A. M. M. critically reviewed, contributed to, and approved the final manuscript.

References

1. Byrne, CD (2010) Fatty liver: role of inflammation and fatty acid nutrition. Prostaglandins Leukot Essent Fatty Acids 82, 265271.CrossRefGoogle Scholar
2. Review, T, LaBrecque, DR, Abbas, Z et al. (2014) World Gastroenterology Organisation global guidelines: nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Clin Gastroenterol 48, 467473.Google Scholar
3. Schuppan, D, Gorrell, MD, Klein, T et al. (2010) The challenge of developing novel pharmacological therapies for non-alcoholic steatohepatitis. Liver Int 30, 795808.CrossRefGoogle ScholarPubMed
4. Milic, S, Lulic, D & Stimac, D (2014) Non-alcoholic fatty liver disease and obesity: biochemical, metabolic and clinical presentations. World J Gastroenterol 20, 93309337.Google Scholar
5. Stojsavljevic, S, Gomercic Palcic, M, Virovic Jukic, L et al. (2014) Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World J Gastroenterol 20, 1807018091.Google Scholar
6. Bhatia, LS, Curzen, NP, Calder, PC et al. (2012) Non-alcoholic fatty liver disease: a new and important cardiovascular risk factor? Eur Heart J 33, 11901200.Google Scholar
7. Berlanga, A, Guiu-Jurado, E, Porras, JA et al. (2014) Molecular pathways in non-alcoholic fatty liver disease. Clin Exp Gastroenterol 7, 221239.Google Scholar
8. Kleiner, DE, Brunt, EM, Van Natta, M et al. (2005) Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41, 13131321.Google Scholar
9. Blachier, M, Leleu, H, Peck-Radosavljevic, M et al. (2013) The burden of liver disease in Europe: a review of available epidemiological data. J Hepatol 58, 593608.Google Scholar
10. Gu, Y & Lambert, JD (2013) Modulation of metabolic syndrome-related inflammation by cocoa. Mol Nutr Food Res 57, 948961.Google Scholar
11. Rodriguez-Mateos, A, Vauzour, D, Krueger, CG et al. (2014) Bioavailability, bioactivity and impact on health of dietary flavonoids and related compounds: an update. Arch Toxicol 88, 18031853.CrossRefGoogle ScholarPubMed
12. Tilg, H & Moschen, AR (2008) Insulin resistance, inflammation, and non-alcoholic fatty liver disease. Trends Endocrinol Metab 19, 371379.CrossRefGoogle ScholarPubMed
13. Serviddio, G, Bellanti, F & Vendemiale, G (2013) Free radical biology for medicine: learning from nonalcoholic fatty liver disease. Free Radic Biol Med 65, 952968.Google Scholar
14. Gupte, AA, Lyon, CJ & Hsueh, WA (2013) Nuclear factor (erythroid-derived 2)-like-2 factor (Nrf2), a key regulator of the antioxidant response to protect against atherosclerosis and nonalcoholic steatohepatitis. Curr Diab Rep 13, 362371.Google Scholar
15. Del Rio, D, Rodriguez-Mateos, A, Spencer, JP et al. (2013) Dietary (poly)phenolics in human health: structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid Redox Signal 18, 18181892.CrossRefGoogle ScholarPubMed
16. Williams, RJ, Spencer, JP & Rice-Evans, C (2004) Flavonoids: antioxidants or signalling molecules? Free Radic Biol Med 36, 838849.CrossRefGoogle ScholarPubMed
17. Gomez-Lechon, MJ, Donato, MT, Martinez-Romero, A et al. (2007) A human hepatocellular in vitro model to investigate steatosis. Chem Biol Interact 165, 106116.CrossRefGoogle ScholarPubMed
18. Vidyashankar, S, Sandeep Varma, R & Patki, PS (2013) Quercetin ameliorate insulin resistance and up-regulates cellular antioxidants during oleic acid induced hepatic steatosis in HepG2 cells. Toxicol In Vitro 27, 945953.Google Scholar
19. Shimada, T, Tokuhara, D, Tsubata, M et al. (2012) Flavangenol (pine bark extract) and its major component procyanidin B1 enhance fatty acid oxidation in fat-loaded models. Eur J Pharmacol 677, 147153.CrossRefGoogle ScholarPubMed
20. Pil Hwang, Y, Gyun Kim, H, Choi, JH et al. (2013) 3-Caffeoyl, 4-dihydrocaffeoylquinic acid from Salicornia herbacea attenuates high glucose-induced hepatic lipogenesis in human HepG2 cells through activation of the liver kinase B1 and silent information regulator T1/AMPK-dependent pathway. Mol Nutr Food Res 57, 471482.Google Scholar
21. Shang, J, Chen, LL, Xiao, FX et al. (2008) Resveratrol improves non-alcoholic fatty liver disease by activating AMP-activated protein kinase. Acta Pharmacol Sin 29, 698706.Google Scholar
22. Liu, JF, Ma, Y, Wang, Y et al. (2011) Reduction of lipid accumulation in HepG2 cells by luteolin is associated with activation of AMPK and mitigation of oxidative stress. Phytother Res: PTR 25, 588596.CrossRefGoogle ScholarPubMed
23. Liu, Y, Wang, D, Zhang, D et al. (2011) Inhibitory effect of blueberry polyphenolic compounds on oleic acid-induced hepatic steatosis in vitro. J Agric Food Chem 59, 1225412263.Google Scholar
24. Wu, CH, Lin, MC, Wang, HC et al. (2011) Rutin inhibits oleic acid induced lipid accumulation via reducing lipogenesis and oxidative stress in hepatocarcinoma cells. J Food Sci 76, T65T72.CrossRefGoogle ScholarPubMed
25. Guo, H, Li, D, Ling, W et al. (2011) Anthocyanin inhibits high glucose-induced hepatic mtGPAT1 activation and prevents fatty acid synthesis through PKCzeta. J Lipid Res 52, 908922.CrossRefGoogle ScholarPubMed
26. Jump, DB, Tripathy, S & Depner, CM (2013) Fatty acid-regulated transcription factors in the liver. Annu Rev Nutr 33, 249269.CrossRefGoogle ScholarPubMed
27. Canto, C & Auwerx, J (2010) AMP-activated protein kinase and its downstream transcriptional pathways. Cell Mol Life Sci 67, 34073423.CrossRefGoogle ScholarPubMed
28. Contreras, AV, Torres, N & Tovar, AR (2013) PPAR-alpha as a key nutritional and environmental sensor for metabolic adaptation. Adv Nutr 4, 439452.Google Scholar
29. Masella, R, Di Benedetto, R, Vari, R et al. (2005) Novel mechanisms of natural antioxidant compounds in biological systems: involvement of glutathione and glutathione-related enzymes. J Nutr Biochem 16, 577586.CrossRefGoogle ScholarPubMed
30. Rodriguez-Ramiro, I, Ramos, S, Bravo, L et al. (2012) Procyanidin B2 induces Nrf2 translocation and glutathione S-transferase P1 expression via ERKs and p38-MAPK pathways and protect human colonic cells against oxidative stress. Eur J Nutr 51, 881892.Google Scholar
31. Rodriguez-Ramiro, I, Martin, MA, Ramos, S et al. (2011) Comparative effects of dietary flavanols on antioxidant defences and their response to oxidant-induced stress on Caco2 cells. Eur J Nutr 50, 313322.Google Scholar
32. Jiang, X, Tang, X, Zhang, P et al. (2014) Cyanidin-3-O-beta-glucoside protects primary mouse hepatocytes against high glucose-induced apoptosis by modulating mitochondrial dysfunction and the PI3 K/Akt pathway. Biochem Pharmacol 90, 135144.CrossRefGoogle Scholar
33. Takahashi, Y, Soejima, Y & Fukusato, T (2012) Animal models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Gastroenterol 18, 23002308.Google Scholar
34. Ueno, T, Torimura, T, Nakamura, T et al. (2009) Epigallocatechin-3-gallate improves nonalcoholic steatohepatitis model mice expressing nuclear sterol regulatory element binding protein-1c in adipose tissue. Int J Mol Med 24, 1722.Google Scholar
35. Ji, G, Yang, Q, Hao, J et al. (2011) Anti-inflammatory effect of genistein on non-alcoholic steatohepatitis rats induced by high fat diet and its potential mechanisms. Int Immunopharmacol 11, 762768.Google Scholar
36. Marcolin, E, Forgiarini, LF, Rodrigues, G et al. (2013) Quercetin decreases liver damage in mice with non-alcoholic steatohepatitis. Basic Clin Pharmacol Toxicol 112, 385391.Google Scholar
37. Gomez-Zorita, S, Fernandez-Quintela, A, Macarulla, MT et al. (2012) Resveratrol attenuates steatosis in obese Zucker rats by decreasing fatty acid availability and reducing oxidative stress. Br J Nutr 107, 202210.Google Scholar
38. Joven, J, Espinel, E, Rull, A et al. (2012) Plant-derived polyphenols regulate expression of miRNA paralogs miR-103/107 and miR-122 and prevent diet-induced fatty liver disease in hyperlipidemic mice. Biochim Biophys Acta 1820, 894899.CrossRefGoogle ScholarPubMed
39. Hui, E, Xu, A, Bo Yang, H et al. (2013) Obesity as the common soil of non-alcoholic fatty liver disease and diabetes: role of adipokines. J Diab Investig 4, 413425.Google Scholar
40. Huang, XD, Fan, Y, Zhang, H et al. (2008) Serum leptin and soluble leptin receptor in non-alcoholic fatty liver disease. World J Gastroenterol 14, 28882893.Google Scholar
41. Buechler, C, Wanninger, J & Neumeier, M (2011) Adiponectin, a key adipokine in obesity related liver diseases. World J Gastroenterol 17, 28012811.Google ScholarPubMed
42. Kim, MH, Kang, KS & Lee, YS (2010) The inhibitory effect of genistein on hepatic steatosis is linked to visceral adipocyte metabolism in mice with diet-induced non-alcoholic fatty liver disease. Br J Nutr 104, 13331342.Google Scholar
43. Park, HJ, Jung, UJ, Lee, MK et al. (2013) Modulation of lipid metabolism by polyphenol-rich grape skin extract improves liver steatosis and adiposity in high fat fed mice. Mol Nutr Food Res 57, 360364.Google Scholar
44. Brown, MS & Goldstein, JL (2008) Selective versus total insulin resistance: a pathogenic paradox. Cell Metab 7, 9596.Google Scholar
45. Huang, C, Qiao, X & Dong, B (2011) Neonatal exposure to genistein ameliorates high-fat diet-induced non-alcoholic steatohepatitis in rats. Br J Nutr 106, 105113.Google Scholar
46. Andrade, JM, Paraiso, AF, de Oliveira, MV et al. (2014) Resveratrol attenuates hepatic steatosis in high-fat fed mice by decreasing lipogenesis and inflammation. Nutrition 30, 915919.Google Scholar
47. Tsuruta, Y, Nagao, K, Kai, S et al. (2011) Polyphenolic extract of lotus root (edible rhizome of Nelumbo nucifera) alleviates hepatic steatosis in obese diabetic db/db mice. Lipids Health Dis 10, 202.Google Scholar
48. Pettinelli, P, Del Pozo, T, Araya, J et al. (2009) Enhancement in liver SREBP-1c/PPAR-alpha ratio and steatosis in obese patients: correlations with insulin resistance and n-3 long-chain polyunsaturated fatty acid depletion. Biochim Biophys Acta 1792, 10801086.Google Scholar
49. Salamone, F, Li Volti, G, Titta, L et al. (2012) Moro orange juice prevents fatty liver in mice. World J Gastroenterol 18, 38623868.Google Scholar
50. Vitaglione, P, Morisco, F, Mazzone, G et al. (2010) Coffee reduces liver damage in a rat model of steatohepatitis: the underlying mechanisms and the role of polyphenols and melanoidins. Hepatology 52, 16521661.Google Scholar
51. Alberdi, G, Rodriguez, VM, Macarulla, MT et al. (2013) Hepatic lipid metabolic pathways modified by resveratrol in rats fed an obesogenic diet. Nutrition 29, 562567.Google Scholar
52. Um, JH, Park, SJ, Kang, H et al. (2010) AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes 59, 554563.Google Scholar
53. Beltran-Debon, R, Rull, A, Rodriguez-Sanabria, F et al. (2011) Continuous administration of polyphenols from aqueous rooibos (Aspalathus linearis) extract ameliorates dietary-induced metabolic disturbances in hyperlipidemic mice. Phytomedicine 18, 414424.Google Scholar
54. Viollet, B, Guigas, B, Leclerc, J et al. (2009) AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives. Acta Physiol (Oxf) 196, 8198.Google Scholar
55. Hardie, DG & Pan, DA (2002) Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase. Biochem Soc Trans 30, 10641070.Google Scholar
56. Mills, SE, Foster, DW & McGarry, JD (1983) Interaction of malonyl-CoA and related compounds with mitochondria from different rat tissues. Relationship between ligand binding and inhibition of carnitine palmitoyltransferase I. Biochem J 214, 8391.CrossRefGoogle ScholarPubMed
57. Meakin, PJ, Chowdhry, S, Sharma, RS et al. (2014) Susceptibility of Nrf2-null mice to steatohepatitis and cirrhosis upon consumption of a high-fat diet is associated with oxidative stress, perturbation of the unfolded protein response, and disturbance in the expression of metabolic enzymes but not with insulin resistance. Mol Cell Biol 34, 33053320.Google Scholar
58. Cui, Y, Wang, Q, Li, X et al. (2013) Experimental nonalcoholic fatty liver disease in mice leads to cytochrome p450 2a5 upregulation through nuclear factor erythroid 2-like 2 translocation. Redox Biol 1, 433440.Google Scholar
59. Li, L, Hai, J, Li, Z et al. (2014) Resveratrol modulates autophagy and NF-kappaB activity in a murine model for treating non-alcoholic fatty liver disease. Food Chem Toxicol 63, 166173.CrossRefGoogle Scholar
60. Bujanda, L, Hijona, E, Larzabal, M et al. (2008) Resveratrol inhibits nonalcoholic fatty liver disease in rats. BMC Gastroenterol 8, 40.Google Scholar
61. Bleau, C, Karelis, AD, St-Pierre, DH et al. (2015) Crosstalk between intestinal microbiota, adipose tissue and skeletal muscle as an early event in systemic low-grade inflammation and the development of obesity and diabetes. Diabetes Metab Res Rev 31, 545561.Google Scholar
62. Marcolin, E, San-Miguel, B, Vallejo, D et al. (2012) Quercetin treatment ameliorates inflammation and fibrosis in mice with nonalcoholic steatohepatitis. J Nutr 142, 18211828.Google Scholar
63. Faghihzadeh, F, Adibi, P, Rafiei, R et al. (2014) Resveratrol supplementation improves inflammatory biomarkers in patients with nonalcoholic fatty liver disease. Nutr Res 34, 837843.Google Scholar
64. Chen, S, Zhao, X, Ran, L et al. (2015) Resveratrol improves insulin resistance, glucose and lipid metabolism in patients with non-alcoholic fatty liver disease: a randomized controlled trial. Dig Liver Dis 47, 226232.Google Scholar
65. Chachay, VS, Macdonald, GA, Martin, JH et al. (2014) Resveratrol does not benefit patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 12, 20922103, e2091e2096.Google Scholar
66. Chang, HC, Peng, CH, Yeh, DM et al. (2014) Hibiscus sabdariffa extract inhibits obesity and fat accumulation, and improves liver steatosis in humans. Food Funct 5, 734739.Google Scholar
67. Guo, H, Zhong, R, Liu, Y et al. (2014) Effects of bayberry juice on inflammatory and apoptotic markers in young adults with features of non-alcoholic fatty liver disease. Nutrition 30, 198203.Google Scholar
68. Calabrese, EJ, Mattson, MP & Calabrese, V (2010) Resveratrol commonly displays hormesis: occurrence and biomedical significance. Hum Exp Toxicol 29, 9801015.Google Scholar
69. Reagan-Shaw, S, Nihal, M & Ahmad, N (2008) Dose translation from animal to human studies revisited. FASEB J 22, 659661.Google Scholar
70. Zamora-Ros, R, Andres-Lacueva, C, Lamuela-Raventos, RM et al. (2008) Concentrations of resveratrol and derivatives in foods and estimation of dietary intake in a Spanish population: European prospective investigation into cancer and nutrition (EPIC)-Spain cohort. Br J Nutr 100, 188196.Google Scholar
71. Rothwell, JA, Perez-Jimenez, J, Neveu, V et al. (2013) Phenol-Explorer 3·0: a major update of the Phenol-Explorer database to incorporate data on the effects of food processing on polyphenol content. Database (Oxf) 2013, bat070.Google Scholar
72. Takahashi, Y & Fukusato, T (2014) Histopathology of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Gastroenterol 20, 1553915548.Google Scholar
73. Wree, A, Broderick, L, Canbay, A et al. (2013) From NAFLD to NASH to cirrhosis-new insights into disease mechanisms. Nat Rev Gastroenterol Hepatol 10, 627636.Google Scholar
74. Lee, CH, Kuo, CY, Wang, CJ et al. (2012) A polyphenol extract of Hibiscus sabdariffa L. ameliorates acetaminophen-induced hepatic steatosis by attenuating the mitochondrial dysfunction in vivo and in vitro. Biosci Biotechnol Biochem 76, 646651.Google Scholar
75. Poulsen, MM, Larsen, JO, Hamilton-Dutoit, S et al. (2012) Resveratrol up-regulates hepatic uncoupling protein 2 and prevents development of nonalcoholic fatty liver disease in rats fed a high-fat diet. Nutr Res 32, 701708.Google Scholar
76. Aoun, M, Michel, F, Fouret, G et al. (2010) A polyphenol extract modifies quantity but not quality of liver fatty acid content in high-fat-high-sucrose diet-fed rats: possible implication of the sirtuin pathway. Br J Nutr 104, 17601770.Google Scholar
Figure 0

Fig. 1. Polyphenol structures.

Figure 1

Fig. 2. (Colour online) Possible mechanisms underlying the effect of polyphenols in non-alcoholic fatty liver disease (NAFLD). Polyphenols may prevent cellular damage in hepatocytes associated with NAFLD through different mechanism of action including: (a) reducing de novo lipogenesis through sterol regulatory element-binding protein 1c (SREBP-1c) down-regulation, (b) increasing β-fatty acid (FA) oxidation by PPARα up-regulation, (c) improving insulin sensitivity (d) reducing oxidative stress through increasing the antioxidant defence levels via nuclear factor-erythroid 2-related factor 2 (Nrf2), (e) attenuating the inflammatory pathways. Presumably SREBP-1c down-regulation and PPARα up-regulation are modulated by AMPK activation (by phosphorylation). TNFR, TNFα receptor; IL6-R, IL-6 receptor; IR, insulin receptor; CD36, cluster of differentiation 36/FA translocase; p-AMPK, phosphorylated AMP-activated protein kinase α; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; SCD, stearoyl-CoA desaturase; GPAT, glycerol-3-phosphate acyltransferase; CPT-1, carnitine palmitoyl transferase 1; ACO, acyl-CoA oxidase; PGC-1, PGC1α, PPARγ coactivator-1α; JNK, c-Jun N-terminal kinase; PKC, protein kinase C; mTOR, mammalian target of rapamycin.

Figure 2

Table 1. Cell culture studies investigating the impact of polyphenols on non-alcoholic fatty liver disease

Figure 3

Table 2. Rodent studies evaluating the impact of polyphenols on non-alcoholic fatty liver disease

Figure 4

Table 3. Clinical trials carried out with polyphenols in non-alcoholic fatty liver disease (NAFLD) subjects