Advertisement
Review Article| Volume 49, ISSUE 1, P63-94, March 2020

Nutrition and Nonalcoholic Fatty Liver Disease

Current Perspectives
Open AccessPublished:December 23, 2019DOI:https://doi.org/10.1016/j.gtc.2019.09.003

      Keywords

      Key points

      • Nonalcoholic fatty liver disease (NAFLD) pathogenesis and progression are complex, heterogeneous, and multifactorial.
      • The human diet typically contains thousands of bioactive molecules that orchestrate a variety of metabolic and signaling processes in health and disease, comprising a natural combination approach.
      • Although food composition varies widely, cumulative evidence suggests that specific dietary macronutrients and micronutrients can affect biological processes involved in NAFLD pathogenesis.

      Obesity is a common feature of many metabolic diseases, including nonalcoholic fatty liver disease

      Obesity was rare in ancient times, often praised and limited to the aristocracy. However, it was sometimes noted that excess body weight could lead to ill health. Hippocrates, in 400 bc, was one of the first to comment on the perils of obesity:“It is injurious to health to take in more food than the constitution will bear, when at the same time one uses no exercise to carry off this excess…”—Translated from Hippocrates, circa 400 bc.

      Hippocrates. De Prisca Medicina. 400 BC.

      Several famous physicians have cataloged the ill health associated with obesity. The anatomist Joannes Morgagni
      • Morgagni J.B.
      Epistola Anatoma Clinica XXI.
      was the first to describe ectopic fat from dissected postmortem studies and associate it with the development of atherosclerosis. William Osler
      • Osler W.
      Lectures on angina pectoris and allied states.
      listed ectopic fat as a causative factor of angina pectoris. By the early nineteenth century, much of the modern understanding of ectopic fat within key organs was being delineated. Although Hippocrates recognized exercise as a mitigatory factor against obesity, Ludovicus Nonnius was one of the first physicians to embrace diet as an important factor in health.
      • Nonni L.
      Diaeteticon, sive, De re cibaria libri IV.
      Thus, the foundations for the implications of obesity/ectopic fat and potential treatments were laid nearly 500 years ago.
      Healthy eating habits, weight loss, and increased physical activity (PA) constitute the cornerstones of the management of nonalcoholic fatty liver disease (NAFLD).
      • Chalasani N.
      • Younossi Z.
      • Lavine J.E.
      • et al.
      The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases.
      ,
      • Romero-Gomez M.
      • Zelber-Sagi S.
      • Trenell M.
      Treatment of NAFLD with diet, physical activity and exercise.
      This article summarizes the scientific evidence for the effects of diet on NAFLD. It also provides a perspective on how certain nutrients in food signal and regulate key molecular pathways implicated in NAFLD pathogenesis. This perspective is not intended to be a comprehensive review of every nutrient; there are many excellent references cited here that provide more in-depth information.
      • Dongiovanni P.
      • Valenti L.
      A nutrigenomic approach to non-alcoholic fatty liver disease.
      • Juarez-Hernandez E.
      • Chavez-Tapia N.C.
      • Uribe M.
      • et al.
      Role of bioactive fatty acids in nonalcoholic fatty liver disease.
      • Marchesini G.
      • Petta S.
      • Dalle Grave R.
      Diet, weight loss, and liver health in nonalcoholic fatty liver disease: pathophysiology, evidence, and practice.
      • McCarthy E.M.
      • Rinella M.E.
      The role of diet and nutrient composition in nonalcoholic Fatty liver disease.
      Based on the current evidence, this article provides a dietary framework that could form an integral part of a comprehensive management strategy for NAFLD.

      Consequences and challenges of caloric imbalance

      A minority of people were obese until recent history, and it is only in the last 50 years that obesity has taken on epidemic proportions. In 2016, more than 1.9 billion adults (39% of the adult population) were overweight (body mass index [BMI] 25.0 to <30 kg/m2), and 650 million (13% of the adult population) were considered obese (BMI ≥ 30.0 kg/m2). Associated with high obesity prevalance are the sequelae of obesity-related complications: NAFLD, metabolic syndrome, type 2 diabetes (T2D), and cardiovascular disease (CVD). NAFLD is rapidly becoming the most important cause of liver disease worldwide, with an estimated global prevalence of approximately 24%.
      • McKay A.
      • Wilman H.R.
      • Dennis A.
      • et al.
      Measurement of liver iron by magnetic resonance imaging in the UK Biobank population.
      ,
      • Younossi Z.M.
      The epidemiology of nonalcoholic steatohepatitis.
      Both T2D and CVD are also effect modifiers of NAFLD; overall prevalence of NAFLD in patients with T2D nearly triples (∼60%), and CVD is the primary cause of death in patients with NAFLD.
      • Dai W.
      • Ye L.
      • Liu A.
      • et al.
      Prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: a meta-analysis.
      ,
      • Allen A.M.
      • Therneau T.M.
      • Larson J.J.
      • et al.
      Nonalcoholic fatty liver disease incidence and impact on metabolic burden and death: a 20 year-community study.
      The obesity epidemic and its sequelae also affect children and adolescents. Hepatic steatosis prevalence among children and adolescents within the European Union was found to be ∼28% (∼1.5 million), with nearly one-fifth of obese children and adolescents having significant acquired cardiovascular risk factors before adulthood, and 4.6% having metabolic syndrome and increased risk of progression to heart disease.
      • Lobstein T.
      • Jackson-Leach R.
      Estimated burden of paediatric obesity and co-morbidities in Europe. Part 2. Numbers of children with indicators of obesity-related disease.
      In the United States, hepatic steatosis was found in 9.6% of individuals aged 2 to 19 years and in 38% of obese children autopsied between 1993 and 2003.
      • Schwimmer J.B.
      • Deutsch R.
      • Kahen T.
      • et al.
      Prevalence of fatty liver in children and adolescents.
      The current recommendation for adults from United Kingdom’s Scientific Advisory Committee on Nutrition is to consume a diet of 50% carbohydrate (CHO) and less than 35% fat of the daily total energy intake (TEI), in conjunction with 0.75 g of protein per kilogram of body weight.
      Public Health England
      Government dietary recommendations: government recommendations for energy and nutrients for males and females aged 1–18 years and 19+ years.
      The recommended daily macronutrient intake and the actual daily intake reported from a cohort of 210,106 participants from the UK Biobank database are presented in Table 1. Twenty-four-hour dietary recall questionnaires revealed that 32% of men and 42% of women were consuming more energy than the recommended daily amount, although the average of the cohort was at or less than the recommended TEI.
      • Bennett E.
      • Peters S.A.E.
      • Woodward M.
      Sex differences in macronutrient intake and adherence to dietary recommendations: findings from the UK Biobank.
      Although women on average exceeded the recommended intake of total sugar, both genders exceeded recommended saturated fat intake, were at less than the recommended levels for polyunsaturated fat intake, and significantly deficient in dietary fiber intake (see Table 1). These dietary findings likely present a best-case scenario because people generally over-report healthy foods and under-report unhealthy foods, and the population sampled in the UK Biobank is considered to be generally healthy.
      • Bennett E.
      • Peters S.A.E.
      • Woodward M.
      Sex differences in macronutrient intake and adherence to dietary recommendations: findings from the UK Biobank.
      Thus, it can be surmised that a more realistic picture of a typical Western diet would include inadequate and/or imbalanced nutrient profiles.
      Table 1United Kingdom recommended daily macronutrient intake and actual intake in a cohort of 210,106 generally healthy participants from the UK Biobank
      Modified from Bennett E, Peters SAE, Woodward M. Sex differences in macronutrient intake and adherence to dietary recommendations: findings from the UK Biobank. BMJ Open. 2018;8(4):e020017; with permission.
      Recommended Daily IntakeActual Daily Intake
      Reported from 24-hour dietary recall questionnaires.
      TEI (kJ)
       Men<10,4609525
       Women<83638168
      Total CHO, TEI (%)
       Men>5049 (271 g)
       Women>5047 (237 g)
      Total Sugars (g)
       Men<120125 (26% TEI)
       Women<90116 (24% TEI)
      Total Fat, TEI (%)
       Men<3532 (83 g)
       Women<3533 (73 g)
      Saturated Fat, TEI (%)
       Men<1112 (32 g)
       Women<1112 (28 g)
      Polyunsaturated Fat, TEI (%)
       Men6–116 (15 g)
       Women6–116 (14 g)
      Fiber (g)
       Men≥3017
       Women≥3016
      Protein (g/kg BW)
       Men0.751.04
       Women1.13
      Abbreviation: BW, body weight.
      a Reported from 24-hour dietary recall questionnaires.
      Although there are inherited conditions associated with excessive body weight, such as congenital leptin deficiency,
      • Montague C.T.
      • Farooqi I.S.
      • Whitehead J.P.
      • et al.
      Congenital leptin deficiency is associated with severe early-onset obesity in humans.
      melanin concentrating hormone receptor-1 deficiency,
      • Wermter A.K.
      • Reichwald K.
      • Buch T.
      • et al.
      Mutation analysis of the MCHR1 gene in human obesity.
      or Prader-Willi syndrome,
      • Bray G.A.
      • Dahms W.T.
      • Swerdloff R.S.
      • et al.
      The Prader-Willi syndrome: a study of 40 patients and a review of the literature.
      in most cases the primary cause of obesity is energy imbalance. The obesity epidemic can be attributed primarily to the increasing propensity for a sedentary lifestyle and the ready availability of food, especially energy-dense foods.
      • Hruby A.
      • Hu F.B.
      The epidemiology of obesity: a big picture.
      Measures to modulate energy homeostasis and nutrient balance remain woefully inadequate. Consequently, interventions that treat obesity and ectopic fat, particularly behavioral modifications to enhance PA and support healthy diet, are of great interest to the clinical and research communities.

      Nonalcoholic fatty liver disease pathogenesis

      During the past decade, extensive multidisciplinary efforts have contributed to a deeper understanding of the complex pathophysiology of the NAFLD spectrum. Broadly, the disease seems to progress from fat accumulation (ie, steatosis or nonalcoholic fatty liver [NAFL]) to inflammation (ie, nonalcoholic steatohepatitis [NASH]) to fibrosis and eventually cirrhosis.
      • Cohen J.C.
      • Horton J.D.
      • Hobbs H.H.
      Human fatty liver disease: old questions and new insights.
      ,
      • Machado M.V.
      • Diehl A.M.
      Pathogenesis of nonalcoholic steatohepatitis.
      Two core principles on the critical nodes of NAFLD pathogenesis have emerged in the context of metabolic substrate overload:
      • 1.
        Disease establishment node: insulin resistance and lipotoxicity.
        Ectopic fat depots within muscle and liver that commonly occur with obesity can contribute to metabolic inflexibility, the inability to adequately regulate fuel substrates (glucose and free fatty acids [FFAs]).
        • Galgani J.E.
        • Moro C.
        • Ravussin E.
        Metabolic flexibility and insulin resistance.
        Metabolic inflexibility results in insulin resistance and contributes to mitochondrial dysfunction, affecting lipid metabolism (Chakravarthy M.V., Neuschwander-Tetri B.A. Submitted for publication). Perturbation of metabolic processes leads to accumulation of intracellular triglycerides and accelerated lipolysis (secondary to adipose tissue insulin resistance), releasing FFAs, which are the precursors of lipotoxic molecules (eg, ceramides, sphingomyelins, lysophospholipids, and others) that cause cellular damage.
        • Mota M.
        • Banini B.A.
        • Cazanave S.C.
        • et al.
        Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease.
        Thus, any treatment approach (dietary or pharmacologic) for NAFLD should consider the underlying lipotoxic environment as a key interventional node.
      • 2.
        Disease progression node: lipotoxic fatty acids (FAs) lead to cellular damage, inflammation, and fibrogenesis.
        Current evidence indicates that the degree of liver fibrosis is the prognostic marker that is most directly correlated to eventual morbidity and mortality in patients with NASH.
        • Angulo P.
        • Kleiner D.E.
        • Dam-Larsen S.
        • et al.
        Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease.
        ,
        • Ekstedt M.
        • Hagstrom H.
        • Nasr P.
        • et al.
        Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up.
        Processes that lead to fibroinflammatory disease have been an intense area of research that has shed light on the pathways that control apoptosis, autophagy, endoplasmic reticulum (ER) stress, tissue repair mechanisms, gut barrier function, and fibrogenesis.
        • Machado M.V.
        • Diehl A.M.
        Pathogenesis of nonalcoholic steatohepatitis.
        ,
        • Bozaykut P.
        • Sahin A.
        • Karademir B.
        • et al.
        Endoplasmic reticulum stress related molecular mechanisms in nonalcoholic steatohepatitis.
        • Kanda T.
        • Matsuoka S.
        • Yamazaki M.
        • et al.
        Apoptosis and non-alcoholic fatty liver diseases.
        • Mouries J.
        • Brescia P.
        • Silvestri A.
        • et al.
        Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development.
        There is heterogeneity in disease progression, likely caused by differences in genetic, microbiome, diet, and other factors. Thus, another key interventional node for NAFLD is to target core pathways that control progression toward more advanced disease.
      Given the pathophysiology of NAFLD, which involves multiple pathways, it raises the question of whether engaging a single molecular target could be sufficient to adequately treat such a complex disease. NAFLD is unlikely to be explained by a single gene defect; it is more likely to involve simultaneous dysregulation of several biochemical functions mediated through a complex set of molecular networks.
      • Cohen J.C.
      • Horton J.D.
      • Hobbs H.H.
      Human fatty liver disease: old questions and new insights.
      • Machado M.V.
      • Diehl A.M.
      Pathogenesis of nonalcoholic steatohepatitis.
      • Galgani J.E.
      • Moro C.
      • Ravussin E.
      Metabolic flexibility and insulin resistance.
      Thus, it is difficult to identify single molecular targets that could sufficiently treat the full spectrum of this heterogeneous disease. Consequently, the authors propose that multiple biochemical pathways that regulate hepatic metabolism, inflammation, and fibrogenesis need to be considered and simultaneously modulated in NAFLD treatment.

      Rationale for combinatorial approaches

      Multiple lines of evidence have converged to support the notion that NAFLD treatment requires a combination therapy approach from both a biological and patient-centric standpoint owing to the complexity, heterogeneity, and multifactorial pathogenesis of the disease. In the Randomized Global Phase 3 Study to Evaluate the Impact on NASH With Fibrosis of Obeticholic Acid Treatment (REGENERATE) trial, 931 individuals with biopsy-confirmed NASH and significant or severe fibrosis (stages F2 or F3) were randomized to receive OCA 10 mg/d (n = 312), OCA 25 mg/d (n = 308), or placebo (n = 311); placebo-subtracted response in NASH resolution or fibrosis improvement was seen in only 3.7% and 11.2% of patients, respectively, in the 25 mg/d OCA group.
      • Younossi Z.
      • Ratziu V.
      • Loomba R.
      • et al.
      Positive results from REGENERATE: a phase 3 international, randomized, placebo-controlled study evaluating obeticholic acid treatment for NASH.
      Recent data show patients with NAFLD often have multiple comorbidities and are typically maintained on multiple medications. In a study of 95 patients with T2D and NAFLD, polypharmacy (5–9 medications) and hyperpolypharmacy (≥10 medications) were present in 59% and 31% of patients, respectively.
      • Patel P.J.
      • Hayward K.L.
      • Rudra R.
      • et al.
      Multimorbidity and polypharmacy in diabetic patients with NAFLD: implications for disease severity and management.
      Therefore, from a patient-centric standpoint, it is desirable to avoid, to the extent possible, additional polypharmacy by considering options that simultaneously address the multiple mechanisms of NAFLD in an integrated manner, including fixed dose combinations instead of sequential additions of multiple individual agents.
      Although scientific debate about the ideal pharmacologic combination continues, one advantage of dietary approaches is that they naturally combine a variety of nutrient and non-nutrient components. The human diet typically contains thousands of bioactive molecules that orchestrate a variety of metabolic and signaling processes in health and disease.
      • De Angelis M.
      • Garruti G.
      • Minervini F.
      • et al.
      The food-gut human axis: the effects of diet on gut microbiota and metabolome.
      ,
      • Scalbert A.
      • Brennan L.
      • Manach C.
      • et al.
      The food metabolome: a window over dietary exposure.
      In addition, these nutrients also modulate some of the same biological pathways and molecular targets as do pharmacologic agents, as discussed later. When considering a complex disease with an incomplete pathophysiologic understanding and the need for a clean safety profile, dietary approaches may be particularly valuable because they offer holistic benefits and address patient-centered needs. For example, dietary approaches have been shown to have a role in improving not only certain features of the NAFLD spectrum but also the concomitant comorbidities of dyslipidemia, insulin resistance, and cardiovascular risk in a safe manner.
      • Esposito K.
      • Maiorino M.I.
      • Bellastella G.
      • et al.
      A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses.
      • Kastorini C.M.
      • Milionis H.J.
      • Esposito K.
      • et al.
      The effect of Mediterranean diet on metabolic syndrome and its components: a meta-analysis of 50 studies and 534,906 individuals.
      • Sofi F.
      • Casini A.
      Mediterranean diet and non-alcoholic fatty liver disease: new therapeutic option around the corner?.
      • Tong T.Y.
      • Wareham N.J.
      • Khaw K.T.
      • et al.
      Prospective association of the Mediterranean diet with cardiovascular disease incidence and mortality and its population impact in a non-Mediterranean population: the EPIC-Norfolk study.
      This article describes the roles of key dietary macronutrients and micronutrients, reviews the current understanding of how these nutrients engage some of the same molecular targets as current pharmacotherapeutic strategies, and describes how such approaches may be leveraged to affect core pathogenic features of NAFLD.

      Macronutrients and micronutrients in nonalcoholic fatty liver disease

      Nutrients significantly influence disease outcomes across several chronic diseases.
      • Tilman D.
      • Clark M.
      Global diets link environmental sustainability and human health.
      Polyphenols, carotenoids, flavonoids, and terpenoids are known to regulate inflammation, proliferation, apoptosis, and angiogenesis.
      • Kotecha R.
      • Takami A.
      • Espinoza J.L.
      Dietary phytochemicals and cancer chemoprevention: a review of the clinical evidence.
      Dietary and lifestyle modifications have been shown to prevent 30% to 40% of all cancers.
      • Dewar S.L.
      • Porter J.
      The effect of evidence-based nutrition clinical care pathways on nutrition outcomes in adult patients receiving non-surgical cancer treatment: a systematic review.
      ,
      • Donaldson M.S.
      Nutrition and cancer: a review of the evidence for an anti-cancer diet.
      Thus, the ways in which specific dietary constituents affect biological processes to affect health outcomes is an area of active investigation.
      • Dongiovanni P.
      • Valenti L.
      A nutrigenomic approach to non-alcoholic fatty liver disease.
      ,
      • Hesketh J.
      Personalised nutrition: how far has nutrigenomics progressed?.
      ,
      • Veselkov K.
      • Gonzalez G.
      • Aljifri S.
      • et al.
      HyperFoods: machine intelligent mapping of cancer-beating molecules in foods.
      This article briefly reviews the key nutrients (Fig. 1) and their molecular mediators (Fig. 2) that have shown significant activity on the multifactorial biology of NAFLD, including changes in liver fat and fibroinflammation.
      Figure thumbnail gr1
      Fig. 1Interactions of macronutrients and micronutrients and their impact on NAFLD pathogenesis and progression. Balanced and/or appropriate levels of dietary macronutrients and micronutrients can act to slow or halt the progression of NAFLD. An imbalance (eg, high levels of fructose or inadequate amounts of polyunsaturated FAs) can contribute to the pathogenesis and progression of NAFLD, which could lead to further cellular damage, inflammation, and fibrogenesis.
      Figure thumbnail gr2
      Fig. 2Impact of key dietary nutrients on molecular mediators that orchestrate the core NAFLD pathobiological pathways. The central features of NAFLD pathogenesis include metabolic dysregulation (eg, increased steatosis, insulin resistance, and mitochondrial dysfunction), inflammation and apoptosis (eg, ER and oxidative stress, immune cell fates), fibrosis (eg, hepatic stellate cell [HSC] activation), and leaky gut (eg, microbiome, tight junction protein regulation). Carbohydrates, FAs, and amino acids can simultaneously either activate or inhibit the critical nodes of NAFLD pathogenesis. AMPK, 5′ adenosine monophosphate-activated protein kinase; Arg, arginine; BCAA, branched-chain amino acids; Cit, citrulline; Cyp2E1, cytochrome P450 Family 2 Subfamily E Member 1; DNL, de novo lipogenesis; FAO, FA oxidation; FAS, FA synthase; FGF21, fibroblast growth factor 21; GCG, glucagon; Gln, glutamine; GLP1, glucagonlike peptide-1; Gly, glycine; GPR, G protein–coupled receptor; HDAC, histone deacetylase; IL6, interleukin 6; JNK, c-Jun N-terminal kinase; M1, macrophage subset 1; M2, macrophage subset 2; mTOR, mammalian target of rapamycin complex; NF-κB, nuclear factor kappa-light-chain enhancer of activated B cells; PPAR, peroxisome proliferator-activated receptor; PUFA, polyunsaturated FAs; SCFA, short-chain FAs; Ser, serine; SFA, saturated FA; SREBP1c, sterol regulatory element-binding protein 1c; TGFβ, transforming growth factor beta; Th17, T-helper 17 cells; TLR4, toll-like receptor 4; TNFα, tumor necrosis factor alpha; Tregs, regulatory T cells; β-cat, beta-catenin.

      Macronutrients and Nonalcoholic Fatty Liver Disease

      Carbohydrates

      Although the typical human diet is naturally rich in CHOs, a refined CHO diet in the context of caloric excess leads to the deposition of intrahepatic triglyceride (IHTG).
      • Sevastianova K.
      • Santos A.
      • Kotronen A.
      • et al.
      Effect of short-term carbohydrate overfeeding and long-term weight loss on liver fat in overweight humans.
      Moreover, CHO intake contributes to 30% of FFA production in patients with NAFLD compared with 5% in a healthy population.
      • Neuschwander-Tetri B.A.
      Carbohydrate intake and nonalcoholic fatty liver disease.
      Nonetheless, under isocaloric conditions it is the type of CHO ingested and not the proportion of dietary energy from CHO that is most relevant. For example, fructose seems to contribute to IHTG even under isocaloric conditions, particularly in liquid form.
      • Stanhope K.L.
      • Schwarz J.M.
      • Keim N.L.
      • et al.
      Consuming fructose-sweetened, not glucose-sweetened, beverages increases visceral adiposity and lipids and decreases insulin sensitivity in overweight/obese humans.
      Epidemiologic studies show a strong association between a sharp increase in fructose consumption and incidence of NAFLD.
      • Stanhope K.L.
      • Schwarz J.M.
      • Havel P.J.
      Adverse metabolic effects of dietary fructose: results from the recent epidemiological, clinical, and mechanistic studies.
      Controlled studies show detrimental effects on insulin sensitivity and IHTG when fructose intake exceeds 25% of energy requirements.
      • Stanhope K.L.
      • Schwarz J.M.
      • Keim N.L.
      • et al.
      Consuming fructose-sweetened, not glucose-sweetened, beverages increases visceral adiposity and lipids and decreases insulin sensitivity in overweight/obese humans.
      Fructose stimulates de novo lipogenesis, inhibits hepatic lipid oxidation by blocking the activity of peroxisome proliferator-activated receptor alpha (PPARα),
      • Roglans N.
      • Vila L.
      • Farre M.
      • et al.
      Impairment of hepatic Stat-3 activation and reduction of PPARalpha activity in fructose-fed rats.
      and increases fibroblast growth factor 21 (FGF21) in a CHO-response element–binding protein (ChREBP)–dependent manner
      • Iroz A.
      • Montagner A.
      • Benhamed F.
      • et al.
      A specific ChREBP and PPARalpha cross-talk is required for the glucose-mediated FGF21 response.
      even when protein intake is controlled.
      • Lundsgaard A.M.
      • Fritzen A.M.
      • Sjoberg K.A.
      • et al.
      Circulating FGF21 in humans is potently induced by short term overfeeding of carbohydrates.
      Fructose also activates c-Jun N-terminal kinase (JNK)
      • Wei Y.
      • Pagliassotti M.J.
      Hepatospecific effects of fructose on c-jun NH2-terminal kinase: implications for hepatic insulin resistance.
      and the nitrooxidative stress marker cytochrome P450-2E1 (CYP2E1).
      • Cho Y.E.
      • Kim D.K.
      • Seo W.
      • et al.
      Fructose promotes leaky gut, endotoxemia, and liver fibrosis through ethanol-inducible cytochrome P450-2E1-mediated oxidative and nitrative stress.
      Cumulatively, these fructose-driven biochemical changes lead to obesity, steatosis, insulin resistance, inflammation, hepatic fibrosis, and leaky gut (see Fig. 2).
      • Roglans N.
      • Vila L.
      • Farre M.
      • et al.
      Impairment of hepatic Stat-3 activation and reduction of PPARalpha activity in fructose-fed rats.
      ,
      • Cho Y.E.
      • Kim D.K.
      • Seo W.
      • et al.
      Fructose promotes leaky gut, endotoxemia, and liver fibrosis through ethanol-inducible cytochrome P450-2E1-mediated oxidative and nitrative stress.
      ,
      • Sellmann C.
      • Priebs J.
      • Landmann M.
      • et al.
      Diets rich in fructose, fat or fructose and fat alter intestinal barrier function and lead to the development of nonalcoholic fatty liver disease over time.
      However, fructose consumed at a limit that reflects usual whole-food intake (eg, fructose as found in fruits) does not meaningfully contribute to de novo lipogenesis, insulin resistance, or hypertriglyceridemia.
      • Agebratt C.
      • Strom E.
      • Romu T.
      • et al.
      A randomized study of the effects of additional fruit and nuts consumption on hepatic fat content, cardiovascular risk factors and basal metabolic rate.
      • Choo V.L.
      • Viguiliouk E.
      • Blanco Mejia S.
      • et al.
      Food sources of fructose-containing sugars and glycaemic control: systematic review and meta-analysis of controlled intervention studies.
      • Weber K.S.
      • Simon M.C.
      • Strassburger K.
      • et al.
      Habitual fructose intake relates to insulin sensitivity and fatty liver index in recent-onset type 2 diabetes patients and individuals without diabetes.
      In contrast, CHO that is somewhat or completely resistant to digestion is associated with reduced risk of CVD and T2D.
      • Mann J.
      • Cummings J.H.
      • Englyst H.N.
      • et al.
      FAO/WHO scientific update on carbohydrates in human nutrition: conclusions.
      ,
      • Ahmadi S.
      • Mainali R.
      • Nagpal R.
      • et al.
      Dietary polysaccharides in the amelioration of gut microbiome dysbiosis and metabolic diseases.
      Importantly, fermentable polysaccharides such as inulin or pectin are metabolized by gut microbiota to produce short-chain FAs (SCFAs), which not only ameliorate insulin resistance
      • Canfora E.E.
      • Jocken J.W.
      • Blaak E.E.
      Short-chain fatty acids in control of body weight and insulin sensitivity.
      but also inhibit histone deacetylases (HDACs) and activate G protein–coupled receptors (GPR41/43) to induce epigenetic and antiinflammatory effects, respectively, to modulate metabolic disease status (see Fig. 2).
      • Li M.
      • van Esch B.
      • Henricks P.A.J.
      • et al.
      The anti-inflammatory effects of short chain fatty acids on lipopolysaccharide- or tumor necrosis factor alpha-stimulated endothelial cells via activation of GPR41/43 and inhibition of HDACs.
      ,
      • Tan J.
      • McKenzie C.
      • Potamitis M.
      • et al.
      The role of short-chain fatty acids in health and disease.
      Some studies have shown favorable effects of dietary fiber on body composition parameters such as reduced body fat percentage, waist circumference, and BMI,
      • Dreher M.L.
      Role of fiber and healthy dietary patterns in body weight regulation and weight loss.
      including reduced insulin resistance,
      • Canfora E.E.
      • Jocken J.W.
      • Blaak E.E.
      Short-chain fatty acids in control of body weight and insulin sensitivity.
      ,
      • Weickert M.O.
      • Mohlig M.
      • Schofl C.
      • et al.
      Cereal fiber improves whole-body insulin sensitivity in overweight and obese women.
      ,
      • Robertson M.D.
      • Bickerton A.S.
      • Dennis A.L.
      • et al.
      Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism.
      through bacterial species that stimulate production of SCFAs.
      • da Silva S.T.
      • dos Santos C.A.
      • Bressan J.
      Intestinal microbiota; relevance to obesity and modulation by prebiotics and probiotics.
      Studies investigating the impact of dietary fiber on liver fat and other NAFLD features are limited. In 1 study of 43 subjects with prediabetes, fiber intake of 20 g/d did not reduce liver fat levels over 12 weeks compared with the control diet of 10 g/d fiber.
      • Errazuriz I.
      • Dube S.
      • Slama M.
      • et al.
      Randomized controlled trial of a MUFA or fiber-rich diet on hepatic fat in prediabetes.
      Additional studies specifically in patients with NAFLD/NASH are needed to better clarify the role of dietary fiber in this population.

      Fat and fatty acids

      Dietary FAs can modulate the activity of key cell types (eg, hepatocytes, macrophages, hepatic stellate cells) implicated across the NAFLD spectrum.
      • Juarez-Hernandez E.
      • Chavez-Tapia N.C.
      • Uribe M.
      • et al.
      Role of bioactive fatty acids in nonalcoholic fatty liver disease.
      Thus, dietary FAs can facilitate the development, prevention, or reversal of some NAFLD features depending on FA composition, carbon chain length, and the molecular targets they engage.
      • Errazuriz I.
      • Dube S.
      • Slama M.
      • et al.
      Randomized controlled trial of a MUFA or fiber-rich diet on hepatic fat in prediabetes.
      ,
      • Masterton G.S.
      • Plevris J.N.
      • Hayes P.C.
      Review article: omega-3 fatty acids - a promising novel therapy for non-alcoholic fatty liver disease.
      Excessive intake of saturated long-chain FAs found in foods such as animal products promotes oxidative stress, mitochondrial dysfunction, and inflammation (see Fig. 2).
      • Sui Y.H.
      • Luo W.J.
      • Xu Q.Y.
      • et al.
      Dietary saturated fatty acid and polyunsaturated fatty acid oppositely affect hepatic NOD-like receptor protein 3 inflammasome through regulating nuclear factor-kappa B activation.
      Overingestion of saturated FAs promotes fatty liver, impairs insulin signaling, and induces hepatic ER stress, a precursor to hepatocyte cellular dysfunction and apoptosis.
      • Pfaffenbach K.T.
      • Gentile C.L.
      • Nivala A.M.
      • et al.
      Linking endoplasmic reticulum stress to cell death in hepatocytes: roles of C/EBP homologous protein and chemical chaperones in palmitate-mediated cell death.
      • Wang D.
      • Wei Y.
      • Pagliassotti M.J.
      Saturated fatty acids promote endoplasmic reticulum stress and liver injury in rats with hepatic steatosis.
      • Rosqvist F.
      • Kullberg J.
      • Stahlman M.
      • et al.
      Overeating saturated fat promotes fatty liver and ceramides compared to polyunsaturated fat: a randomized trial.
      Saturated FA–induced oxidative stress results in the activation of the JNK pathway, a key mechanism in the pathophysiology of NASH and insulin resistance.
      • Seki E.
      • Brenner D.A.
      • Karin M.
      A liver full of JNK: signaling in regulation of cell function and disease pathogenesis, and clinical approaches.
      In contrast, dietary monounsaturated FAs (MUFAs; eg, oleic acid) and polyunsaturated FAs (eg, linoleic acid [n-6], alpha-linolenic acid [n-3], and arachidonic acid), found in foods such as nuts, olive oil, and avocados, have been shown in some studies to reduce IHTG accumulation and inflammation (see Fig. 2). For example, 12 weeks of an MUFA diet (28% of TEI from MUFA, with 50% of the MUFA from olive oil) in 43 subjects with prediabetes decreased hepatic fat and improved both hepatic and total insulin sensitivity.
      • Errazuriz I.
      • Dube S.
      • Slama M.
      • et al.
      Randomized controlled trial of a MUFA or fiber-rich diet on hepatic fat in prediabetes.
      One gram of daily polyunsaturated FA (n-3) supplementation for 12 months promoted lipid oxidation, ameliorated hepatic steatosis, and improved insulin sensitivity in both adult (n = 56; mean age, 58 years)
      • Capanni M.
      • Calella F.
      • Biagini M.R.
      • et al.
      Prolonged n-3 polyunsaturated fatty acid supplementation ameliorates hepatic steatosis in patients with non-alcoholic fatty liver disease: a pilot study.
      and pediatric (n = 108; mean age, 14 years)
      • Boyraz M.
      • Pirgon O.
      • Dundar B.
      • et al.
      Long-term treatment with n-3 polyunsaturated fatty acids as a monotherapy in children with nonalcoholic fatty liver disease.
      patients with NAFLD. However, the precise roles of the types of polyunsaturated FAs to affect liver-related parameters remain unresolved because their effects are complicated by both the ratio of n-3 to n-6 and the confounding influence of background dietary CHO or protein levels.
      Dietary intake of polyunsaturated FAs in a cross-sectional study of patients with NAFLD showed that more than 80% of patients did not reach the daily recommended intake of linolenic and linoleic acids.
      • Da Silva H.E.
      • Arendt B.M.
      • Noureldin S.A.
      • et al.
      A cross-sectional study assessing dietary intake and physical activity in Canadian patients with nonalcoholic fatty liver disease vs healthy controls.
      Well-controlled human clinical trials show that n-6 polyunsaturated FA (linoleic acid) compared with saturated FA (butter or palm oil) prevents IHTG in the context of 7 weeks of overfeeding (n = 39)
      • Rosqvist F.
      • Iggman D.
      • Kullberg J.
      • et al.
      Overfeeding polyunsaturated and saturated fat causes distinct effects on liver and visceral fat accumulation in humans.
      or 10 weeks of isocaloric balance (n = 67).
      • Bjermo H.
      • Iggman D.
      • Kullberg J.
      • et al.
      Effects of n-6 PUFAs compared with SFAs on liver fat, lipoproteins, and inflammation in abdominal obesity: a randomized controlled trial.
      In addition, administering the n-3 polyunsaturated FA, docosahexaenoic acid, at 250 mg/d for 18 months to 20 children (mean age, 10 years) with biopsy-proven NAFLD significantly improved histologic parameters of steatosis, ballooning, and lobular inflammation, but not fibrosis.
      • Nobili V.
      • Carpino G.
      • Alisi A.
      • et al.
      Role of docosahexaenoic acid treatment in improving liver histology in pediatric nonalcoholic fatty liver disease.
      Patients with NASH had a significantly higher intake of n-6 FAs and a decreased n-6/n-3 ratio versus controls.
      • Cortez-Pinto H.
      • Jesus L.
      • Barros H.
      • et al.
      How different is the dietary pattern in non-alcoholic steatohepatitis patients?.
      This finding may reflect the independent role of n-3s in influencing NASH pathogenesis rather than any detrimental role attributed specifically to n-6. Treatment with glucagon like peptide-1 (GLP-1) analogue (exendin-4) improves steatohepatitis and modulates the hepatic n-3/n-6 ratio, specifically by the regulation of hepatic FA metabolism.
      • Kawaguchi T.
      • Itou M.
      • Taniguchi E.
      • et al.
      Exendin4, a glucagonlike peptide1 receptor agonist, modulates hepatic fatty acid composition and Delta5desaturase index in a murine model of nonalcoholic steatohepatitis.
      FAs are potent endogenous ligands for canonical nutrient-sensed transcription factors, such as PPARs, which alter tissue FA compositions and induce cell-signaling pathways to regulate genes implicated in lipid synthesis and oxidation.
      • Chakravarthy M.V.
      • Lodhi I.J.
      • Yin L.
      • et al.
      Identification of a physiologically relevant endogenous ligand for PPARalpha in liver.
      ,
      • Kim J.H.
      • Song J.
      • Park K.W.
      The multifaceted factor peroxisome proliferator-activated receptor gamma (PPARgamma) in metabolism, immunity, and cancer.
      PPARs preferentially bind to unsaturated FAs, whereas saturated FAs are generally poor PPAR ligands,
      • Xu H.E.
      • Lambert M.H.
      • Montana V.G.
      • et al.
      Molecular recognition of fatty acids by peroxisome proliferator-activated receptors.
      underscoring that it is the type of fat, and not the amount of fat or its caloric value, that modulates PPAR activity and, consequently, hepatic lipid metabolism.
      • Forman B.M.
      • Chen J.
      • Evans R.M.
      Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta.
      ,
      • Krey G.
      • Braissant O.
      • L'Horset F.
      • et al.
      Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay.
      Polyunsaturated FAs specifically inhibit the expression of sterol regulatory element-binding protein 1c (SREBP1c) while inducing FA oxidizing enzymes
      • Nakamura M.T.
      • Cheon Y.
      • Li Y.
      • et al.
      Mechanisms of regulation of gene expression by fatty acids.
      and suppressing nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) via PPARα,
      • Zuniga J.
      • Cancino M.
      • Medina F.
      • et al.
      N-3 PUFA supplementation triggers PPAR-alpha activation and PPAR-alpha/NF-kappaB interaction: anti-inflammatory implications in liver ischemia-reperfusion injury.
      and bind to the FA receptor, GPR120, to mediate anti-inflammatory and insulin-sensitizing effects (see Fig. 2).
      • Oh D.Y.
      • Talukdar S.
      • Bae E.J.
      • et al.
      GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects.

      Protein and amino acids

      Protein:

      Although attention has largely been focused on CHO and fat with respect to their roles in T2D and CVD development, protein has received less attention. This omission is now beginning to change and a growing body of data suggests that a high protein intake could help reduce liver fat levels. Increased dietary protein content has been shown to attenuate the increased IHTG level observed following hypercaloric feeding with fructose or fat.
      • Bortolotti M.
      • Kreis R.
      • Debard C.
      • et al.
      High protein intake reduces intrahepatocellular lipid deposition in humans.
      A prospective study of 37 patients with T2D and NAFLD fed a high-protein diet (30% protein, 40% CHO) showed a 36% to 48% reduction in IHTG level assessed by MRI, regardless of whether the protein came from animal or plant sources and independent of body weight changes.
      • Markova M.
      • Pivovarova O.
      • Hornemann S.
      • et al.
      Isocaloric diets high in animal or plant protein reduce liver fat and inflammation in individuals with type 2 diabetes.
      Adipose tissue insulin resistance index and levels of markers of hepatic necroinflammation were reduced, and serum levels of FGF21 decreased by 50%, the latter significantly correlated with loss of hepatic fat.
      • Markova M.
      • Pivovarova O.
      • Hornemann S.
      • et al.
      Isocaloric diets high in animal or plant protein reduce liver fat and inflammation in individuals with type 2 diabetes.
      In a subset of the PREVIEW (Prevention of Diabetes Through Lifestyle Intervention and Population Studies in Europe and Around the World) cohort, 25 patients with NAFLD who were obese and insulin resistant were administered a weight-maintaining diet containing either 15% or 25% protein for up to 2 years following an initial weight loss period of 8 weeks. Both groups reduced IHTG levels, visceral adipose tissue (VAT) levels, subcutaneous adipose tissue levels, homeostatic model assessment score for insulin resistance (HOMA-IR), and insulin sensitivity index independent of body weight.
      • Drummen M.
      • Dorenbos E.
      • Vreugdenhil A.C.E.
      • et al.
      Long-term effects of increased protein intake after weight loss on intrahepatic lipid content and implications for insulin sensitivity: a PREVIEW study.
      Protein intake (grams per day) at 6 months was inversely correlated to IHTG and VAT levels,
      • Drummen M.
      • Dorenbos E.
      • Vreugdenhil A.C.E.
      • et al.
      Long-term effects of increased protein intake after weight loss on intrahepatic lipid content and implications for insulin sensitivity: a PREVIEW study.
      showing a dose dependent effect of protein. A recent trial combining moderate CHO restriction (30% of calories) with a high-protein diet (30%) reduced the absolute hepatic fat content by 2.4% in adults with T2D compared with a 0.2% increase observed in those on a high-CHO (50%), normal-protein (17%) diet.
      • Skytte M.J.
      • Samkani A.
      • Petersen A.D.
      • et al.
      A carbohydrate-reduced high-protein diet improves HbA1c and liver fat content in weight stable participants with type 2 diabetes: a randomised controlled trial.
      Nevertheless, protein has been linked to insulin resistance. In human studies, a higher-protein (1.2 g/kg/d) versus lower-protein (0.8 g/kg/d) hypocaloric diet attenuated the weight loss–mediated improvement in insulin sensitivity in 34 sedentary, obese, postmenopausal women.
      • Smith G.I.
      • Yoshino J.
      • Kelly S.C.
      • et al.
      High-protein intake during weight loss therapy eliminates the weight-loss-induced improvement in insulin action in obese postmenopausal women.
      Another study by the same group evaluated the effects of a diet consisting of either 0.6 g protein per kilogram fat-free mass (containing 0.0684 g of leucine per kilogram fat-free mass) or leucine matched to protein (0.0684 g leucine per kilogram fat-free mass) in 30 obese, insulin-resistant women with NAFLD.
      • Harris L.L.S.
      • Smith G.I.
      • Patterson B.W.
      • et al.
      Alterations in 3-hydroxyisobutyrate and FGF21 metabolism are associated with protein ingestion-induced insulin resistance.
      Ingestion of protein, but not leucine, decreased insulin-stimulated glucose disposal and prevented both the insulin-mediated decrease in levels of plasma 3-hydroxyisobutyrate, an insulin resistance–inducing valine metabolite,
      • Jang C.
      • Oh S.F.
      • Wada S.
      • et al.
      A branched-chain amino acid metabolite drives vascular fatty acid transport and causes insulin resistance.
      and the increase in FGF21 level.
      • Harris L.L.S.
      • Smith G.I.
      • Patterson B.W.
      • et al.
      Alterations in 3-hydroxyisobutyrate and FGF21 metabolism are associated with protein ingestion-induced insulin resistance.
      Furthermore, preclinical studies
      • Grandison R.C.
      • Piper M.D.
      • Partridge L.
      Amino-acid imbalance explains extension of lifespan by dietary restriction in Drosophila.
      • Piper M.D.
      • Partridge L.
      • Raubenheimer D.
      • et al.
      Dietary restriction and aging: a unifying perspective.
      • Levine M.E.
      • Suarez J.A.
      • Brandhorst S.
      • et al.
      Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population.
      • Solon-Biet S.M.
      • Mitchell S.J.
      • Coogan S.C.
      • et al.
      Dietary protein to carbohydrate ratio and caloric restriction: comparing metabolic outcomes in mice.
      • Fontana L.
      • Cummings N.E.
      • Arriola Apelo S.I.
      • et al.
      Decreased consumption of branched-chain amino acids improves metabolic health.
      • Cummings N.E.
      • Williams E.M.
      • Kasza I.
      • et al.
      Restoration of metabolic health by decreased consumption of branched-chain amino acids.
      suggest that high-protein diets may have negative effects on insulin sensitivity, whereas low-protein diets exert metabolic benefits, supporting the observations noted earlier in humans.
      • Smith G.I.
      • Yoshino J.
      • Kelly S.C.
      • et al.
      High-protein intake during weight loss therapy eliminates the weight-loss-induced improvement in insulin action in obese postmenopausal women.
      ,
      • Harris L.L.S.
      • Smith G.I.
      • Patterson B.W.
      • et al.
      Alterations in 3-hydroxyisobutyrate and FGF21 metabolism are associated with protein ingestion-induced insulin resistance.
      These findings underscore the importance of balancing the quantity and quality of dietary protein relative to other nutrients as a key determinant of metabolic health.
      • Piper M.D.W.
      • Soultoukis G.A.
      • Blanc E.
      • et al.
      Matching dietary amino acid balance to the in silico-translated exome optimizes growth and reproduction without cost to lifespan.
      Certain amino acids in the diet may preferentially modulate key biological processes. For instance, recent studies in flies reported that, when specific dietary amino acids are matched to protein-coding genes, growth and reproduction are optimized without affecting lifespan.
      • Piper M.D.W.
      • Soultoukis G.A.
      • Blanc E.
      • et al.
      Matching dietary amino acid balance to the in silico-translated exome optimizes growth and reproduction without cost to lifespan.
      Specific dietary amino acids have also been shown to modulate several aspects of NAFLD pathogenesis, including glucose homeostasis, fibroinflammation, and gut epithelial barrier integrity, as summarized here.

      Branched-chain amino acids:

      Leucine, isoleucine, and valine (together termed branched-chain amino acids [BCAAs]) regulate several important hepatic metabolic signaling pathways, including insulin signaling, glucose regulation, and efficient channeling of carbon substrates for oxidation through mitochondrial tricarboxylic acid (TCA) cycle. An impaired BCAA-mediated upregulation of the TCA cycle is thought to be a core defect resulting in mitochondrial dysfunction in NAFLD.
      • Sunny N.E.
      • Kalavalapalli S.
      • Bril F.
      • et al.
      Cross-talk between branched-chain amino acids and hepatic mitochondria is compromised in nonalcoholic fatty liver disease.
      High plasma BCAA levels, commonly seen in patients with T2D and insulin-resistant NAFLD/NASH,
      • Iwasa M.
      • Ishihara T.
      • Mifuji-Moroka R.
      • et al.
      Elevation of branched-chain amino acid levels in diabetes and NAFL and changes with antidiabetic drug treatment.
      may not only reflect abnormal glucose metabolism but also increased muscle protein breakdown coupled with downregulation of BCAA catabolizing enzymes, all of which may represent an adaptive physiologic response to hepatic stress in patients with NAFLD.
      • Lake A.D.
      • Novak P.
      • Shipkova P.
      • et al.
      Branched chain amino acid metabolism profiles in progressive human nonalcoholic fatty liver disease.
      BCAA supplementation has been shown to improve steatosis, plasma lipid levels, and glucose tolerance in both rodent NASH models and in patients with NASH-related cirrhosis.
      • Honda T.
      • Ishigami M.
      • Luo F.
      • et al.
      Branched-chain amino acids alleviate hepatic steatosis and liver injury in choline-deficient high-fat diet induced NASH mice.
      • Li T.
      • Geng L.
      • Chen X.
      • et al.
      Branched-chain amino acids alleviate nonalcoholic steatohepatitis in rats.
      • Miyake T.
      • Abe M.
      • Furukawa S.
      • et al.
      Long-term branched-chain amino acid supplementation improves glucose tolerance in patients with nonalcoholic steatohepatitis-related cirrhosis.
      Improvements in steatosis, inflammation, and fibrosis in NASH mouse models seem to be mediated via activation of mammalian target of rapamycin complex 1 (mTORC-1), inhibition of hepatic lipogenic enzymes such as FA synthase (FAS), and via transforming growth factor β (TGFβ)–mediated and Wnt/β-catenin–signaling pathways (see Fig. 2).
      • Takegoshi K.
      • Honda M.
      • Okada H.
      • et al.
      Branched-chain amino acids prevent hepatic fibrosis and development of hepatocellular carcinoma in a non-alcoholic steatohepatitis mouse model.

      Glutamine-serine-glycine axis:

      Levels of glutamine, serine, and glycine, which collectively affect glutathione synthesis, inflammation, and oxidative stress pathways, are also altered in NAFLD. A glutamate-serine-glycine index (glutamate/[serine + glycine]) was correlated with hepatic insulin resistance and γ-glutamyltransferase, independent of BMI, and was able to discriminate fibrosis F3 to F4 from F0 to F2.
      • Gaggini M.
      • Carli F.
      • Rosso C.
      • et al.
      Altered amino acid concentrations in NAFLD: impact of obesity and insulin resistance.
      Glutamine is the most abundant amino acid in both the intracellular and extracellular compartments, and was found to be critical to maintaining intestinal mucosal integrity
      • Panigrahi P.
      • Gewolb I.H.
      • Bamford P.
      • et al.
      Role of glutamine in bacterial transcytosis and epithelial cell injury.
      ,
      • DeMarco V.G.
      • Li N.
      • Thomas J.
      • et al.
      Glutamine and barrier function in cultured Caco-2 epithelial cell monolayers.
      by regulating epithelial tight junction proteins.
      • Basuroy S.
      • Sheth P.
      • Mansbach C.M.
      • et al.
      Acetaldehyde disrupts tight junctions and adherens junctions in human colonic mucosa: protection by EGF and L-glutamine.
      Macrophage activation is considered a critical event for NASH progression, and glutamine is necessary to maintain the anti-inflammatory alternative activation pathway in macrophages.
      • Jha A.K.
      • Huang S.C.
      • Sergushichev A.
      • et al.
      Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization.
      ,
      • Liu P.S.
      • Wang H.
      • Li X.
      • et al.
      alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming.
      Macrophages depleted of glutamine express a proinflammatory transcriptome and phenocopy PPARγ-deficient unstimulated macrophages, indicating the requirement of PPARγ for glutamine metabolism (see Fig. 2).
      • Nelson V.L.
      • Nguyen H.C.B.
      • Garcia-Canaveras J.C.
      • et al.
      PPARgamma is a nexus controlling alternative activation of macrophages via glutamine metabolism.
      Serine deficiency, observed in patients with NASH,
      • Mardinoglu A.
      • Agren R.
      • Kampf C.
      • et al.
      Genome-scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non-alcoholic fatty liver disease.
      ,
      • Mardinoglu A.
      • Bjornson E.
      • Zhang C.
      • et al.
      Personal model-assisted identification of NAD(+) and glutathione metabolism as intervention target in NAFLD.
      was shown to directly affect cellular acylcarnitine levels, a signature of altered mitochondrial function and, consequently, impaired fuel use from FAs leading to mitochondrial fragmentation.
      • Gao X.
      • Lee K.
      • Reid M.A.
      • et al.
      Serine availability influences mitochondrial dynamics and function through lipid metabolism.
      In a detailed metabolomic study, enzymatic expression and DNA methylation analyses were performed in a high-fat, high-fructose fed NAFLD murine model that showed 30% depletion of hepatic methionine, whereas s-adenosylhomocysteine and homocysteine were significantly increased (25%–35%) and serine, a substrate for both homocysteine remethylation and transsulfuration, was depleted during NASH development.
      • Pacana T.
      • Cazanave S.
      • Verdianelli A.
      • et al.
      Dysregulated hepatic methionine metabolism drives homocysteine elevation in diet-induced nonalcoholic fatty liver disease.
      In contrast, serine supplementation in high-fat fed mice increased insulin sensitivity and reduced hepatic lipid accumulation without affecting body weight by epigenetic modulation of glutathione synthesis–related genes through 5′ adenosine monophosphate–activated protein kinase (AMPK) activation (see Fig. 2).
      • Zhou X.
      • He L.
      • Zuo S.
      • et al.
      Serine prevented high-fat diet-induced oxidative stress by activating AMPK and epigenetically modulating the expression of glutathione synthesis-related genes.
      Serine supplementation of 20 g/d for 14 days in a small (n = 6) cohort of patients with NAFLD decreased serum alanine aminotransferase (ALT) level and improved hepatic steatosis.
      • Mardinoglu A.
      • Bjornson E.
      • Zhang C.
      • et al.
      Personal model-assisted identification of NAD(+) and glutathione metabolism as intervention target in NAFLD.
      Glycine supplementation in preclinical models was found to exert antiinflammatory, immunomodulatory, cytoprotective, platelet-stabilizing, and antiangiogenic effects in part mediated by blunted activation of p38 mitogen-activated protein kinase and JNK and decreased Fas ligand expression (see Fig. 2).
      • Zhong X.
      • Li X.
      • Qian L.
      • et al.
      Glycine attenuates myocardial ischemia-reperfusion injury by inhibiting myocardial apoptosis in rats.
      ,
      • McCarty M.F.
      • Barroso-Aranda J.
      • Contreras F.
      The hyperpolarizing impact of glycine on endothelial cells may be anti-atherogenic.
      Lower glycine levels were significantly associated with increasing numbers of metabolic syndrome components in a population-based, cross-sectional survey of 472 Chinese individuals.
      • Li X.
      • Sun L.
      • Zhang W.
      • et al.
      Association of serum glycine levels with metabolic syndrome in an elderly Chinese population.
      Emerging evidence also suggests that glycine has a unique ability to stimulate secretion of both GLP-1 and glucagon (GCG),
      • Gameiro A.
      • Reimann F.
      • Habib A.M.
      • et al.
      The neurotransmitters glycine and GABA stimulate glucagon-like peptide-1 release from the GLUTag cell line.
      • Rubio I.G.
      • Castro G.
      • Zanini A.C.
      • et al.
      Oral ingestion of a hydrolyzed gelatin meal in subjects with normal weight and in obese patients: postprandial effect on circulating gut peptides, glucose and insulin.
      • Gannon M.C.
      • Nuttall J.A.
      • Nuttall F.Q.
      The metabolic response to ingested glycine.
      mimicking the actions of the native hormone oxyntomodulin, which is released from intestinal L cells in response to meals and activates both the GLP-1 and GCG receptors.
      • Baldissera F.G.
      • Holst J.J.
      • Knuhtsen S.
      • et al.
      Oxyntomodulin (glicentin-(33-69)): pharmacokinetics, binding to liver cell membranes, effects on isolated perfused pig pancreas, and secretion from isolated perfused lower small intestine of pigs.
      The pharmacologic unimolecular dual GLP-1/GCG coagonist has been shown to induce a 15% to 20% weight loss, modestly decrease food intake, while boosting thermogenesis, decreasing hepatic and serum triglyceride and cholesterol levels, improving insulin sensitivity, and counteracting leptin resistance in diet-induced obese mice and nonhuman primates.
      • Pocai A.
      • Carrington P.E.
      • Adams J.R.
      • et al.
      Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice.
      ,
      • Henderson S.J.
      • Konkar A.
      • Hornigold D.C.
      • et al.
      Robust anti-obesity and metabolic effects of a dual GLP-1/glucagon receptor peptide agonist in rodents and non-human primates.
      In a randomized, double-blinded crossover study, coinfusion of GLP-1 (0.8 pmol/kg/min) and GCG (50 ng/kg/min) increased energy expenditure and decreased food intake in obese volunteers,
      • Tan T.M.
      • Field B.C.
      • McCullough K.A.
      • et al.
      Coadministration of glucagon-like peptide-1 during glucagon infusion in humans results in increased energy expenditure and amelioration of hyperglycemia.
      underscoring a key reason for the many coagonists currently being evaluated in clinical trials.
      A glycine-induced dual action on GLP-1 and GCG in the liver may explain the observations of glycine supplementation to counteract the fructose-mediated increase in IHTG level.
      • Zhou X.
      • Han D.
      • Xu R.
      • et al.
      Glycine protects against high sucrose and high fat-induced non-alcoholic steatohepatitis in rats.
      ,
      • McCarty M.F.
      • DiNicolantonio J.J.
      The cardiometabolic benefits of glycine: is glycine an 'antidote' to dietary fructose?.
      Although glycine supplementation did not induce weight loss or suppress calorie intake in sucrose-fed mice, it reduced visceral fat stores by more than 50%, increased thermogenic potential of hepatic mitochondria by increasing state 4 respiration, alleviated hepatic steatosis, and improved insulin sensitivity and serum lipid levels (see Fig. 2),
      • Day J.W.
      • Ottaway N.
      • Patterson J.T.
      • et al.
      A new glucagon and GLP-1 co-agonist eliminates obesity in rodents.
      phenocopying some of the effects observed with the coagonist drugs.

      Arginine and citrulline:

      Dietary arginine and citrulline seem to affect gut epithelial barrier integrity, a process increasingly recognized as a core pathogenic feature of NASH progression.
      • Mouries J.
      • Brescia P.
      • Silvestri A.
      • et al.
      Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development.
      ,
      • Rahman K.
      • Desai C.
      • Iyer S.S.
      • et al.
      Loss of junctional adhesion molecule A promotes severe steatohepatitis in mice on a diet high in saturated fat, fructose, and cholesterol.
      Western-style diet–fed mice were supplemented separately with arginine and citrulline. Both amino acids preserved tight junction protein levels in duodenum and decreased bacterial endotoxin in portal plasma and hepatic toll-like receptor 4 (TLR4) messenger RNA, underscoring their importance in maintaining intestinal immunohomeostasis.
      • Sellmann C.
      • Degen C.
      • Jin C.J.
      • et al.
      Oral arginine supplementation protects female mice from the onset of non-alcoholic steatohepatitis.
      ,
      • Sellmann C.
      • Jin C.J.
      • Engstler A.J.
      • et al.
      Oral citrulline supplementation protects female mice from the development of non-alcoholic fatty liver disease (NAFLD).
      Citrulline-fed mice also had decreased plasma proinflammatory cytokine (interleukin 6 [IL6] and tumor necrosis factor alpha [TNFα]) levels and improved plasma triglyceride and insulin levels, whereas in the colon they had decreased TNFα and TLR4 gene expression, increased tight junction protein (claudin-1) levels, and induced growth of the gut-protective Bacteroides/Prevotella (see Fig. 2).
      • Jegatheesan P.
      • Beutheu S.
      • Freese K.
      • et al.
      Preventive effects of citrulline on Western diet-induced non-alcoholic fatty liver disease in rats.
      Recent studies also suggest a central role for arginine in modulating the nitric oxide pathway, critical in the maintenance of gut mucosal integrity in an inducible nitric oxide synthase–dependent manner
      • Coburn L.A.
      • Gong X.
      • Singh K.
      • et al.
      L-arginine supplementation improves responses to injury and inflammation in dextran sulfate sodium colitis.
      and metabolism of energy substrates by stimulating mitochondrial biogenesis.
      • Jobgen W.S.
      • Fried S.K.
      • Fu W.J.
      • et al.
      Regulatory role for the arginine-nitric oxide pathway in metabolism of energy substrates.
      Clinically, arginine supplementation (8.3 g/d for 21 days) in a group of 16 obese, insulin-resistant patients with T2D improved glucose homeostasis, waist circumference, blood pressure, lipid parameters, and endothelial function
      • Lucotti P.
      • Setola E.
      • Monti L.D.
      • et al.
      Beneficial effects of a long-term oral L-arginine treatment added to a hypocaloric diet and exercise training program in obese, insulin-resistant type 2 diabetic patients.
      ; however, effects of dietary arginine on specific liver-related parameters remain to be determined in patients with NAFLD/NASH.

      Fibroblast growth factor 21, sensor of dietary protein status and cellular stress:

      Although protein and amino acids in food induce a plethora of cellular effects, multiple lines of evidence have converged to support FGF21 as a critical sensor of dietary protein and amino acid status. FGF21, a liver-derived metabolic hormone, is robustly induced by the restriction of dietary protein or amino acids, including leucine, methionine, cysteine, asparagine, and several other nonessential amino acids, regardless of the CHO content or total caloric load of the diet.
      • De Sousa-Coelho A.L.
      • Marrero P.F.
      • Haro D.
      Activating transcription factor 4-dependent induction of FGF21 during amino acid deprivation.
      • Wanders D.
      • Forney L.A.
      • Stone K.P.
      • et al.
      FGF21 mediates the thermogenic and insulin-sensitizing effects of dietary methionine restriction but not its effects on hepatic lipid metabolism.
      • Wanders D.
      • Stone K.P.
      • Dille K.
      • et al.
      Metabolic responses to dietary leucine restriction involve remodeling of adipose tissue and enhanced hepatic insulin signaling.
      • Maida A.
      • Zota A.
      • Sjoberg K.A.
      • et al.
      A liver stress-endocrine nexus promotes metabolic integrity during dietary protein dilution.
      In contrast, in protein-replete states (eg, 30% protein), FGF21 was shown to be markedly suppressed.
      • Markova M.
      • Pivovarova O.
      • Hornemann S.
      • et al.
      Isocaloric diets high in animal or plant protein reduce liver fat and inflammation in individuals with type 2 diabetes.
      FGF21 therefore responds to a nutritional state that is different from leptin and other energy balance signals coordinating the adaptive behavioral and metabolic responses to protein restriction.
      • Hill C.M.
      • Berthoud H.R.
      • Munzberg H.
      • et al.
      Homeostatic sensing of dietary protein restriction: a case for FGF21.
      Various manipulations that trigger cellular stress increase FGF21 production, even in tissues such as muscle that normally do not produce significant quantities of FGF21, via the activating transcription factor 4 (ATF4)–dependent pathway,
      • Jiang S.
      • Yan C.
      • Fang Q.C.
      • et al.
      Fibroblast growth factor 21 is regulated by the IRE1alpha-XBP1 branch of the unfolded protein response and counteracts endoplasmic reticulum stress-induced hepatic steatosis.
      ,
      • Schaap F.G.
      • Kremer A.E.
      • Lamers W.H.
      • et al.
      Fibroblast growth factor 21 is induced by endoplasmic reticulum stress.
      and strongly reflect liver fat accumulation and dysregulation of PPARα signaling.
      • Rusli F.
      • Deelen J.
      • Andriyani E.
      • et al.
      Fibroblast growth factor 21 reflects liver fat accumulation and dysregulation of signalling pathways in the liver of C57BL/6J mice.
      ATF4 is a key molecular mediator of the classic integrated stress response, which coordinates the cellular response to various stressors via activation of the amino acid sensor GCN2 (general control nonderepressible 2), increased eIF2a (eukaryotic translation initiation factor 2a) phosphorylation, and subsequent binding of ATF4 to amino acid response elements on the FGF21 promoter, which increases FGF21 expression.
      • Wek R.C.
      • Jiang H.Y.
      • Anthony T.G.
      Coping with stress: eIF2 kinases and translational control.
      ,
      • Kilberg M.S.
      • Shan J.
      • Su N.
      ATF4-dependent transcription mediates signaling of amino acid limitation.
      Thus, connection between hepatic FGF21 secretion and obesity, steatohepatitis, and metabolic stress has led to a broader view of FGF21, specifically its increase, as a signal of both nutrient (protein/amino acid restriction) and cellular (oxidative/ER) stress (see Fig. 2).
      • Maratos-Flier E.
      Fatty liver and FGF21 physiology.

      Micronutrients and Nonalcoholic Fatty Liver Disease

      Choline

      Choline is metabolized largely in the liver into phosphatidylcholine and plays an important role in the assembly and secretion of lipoproteins and the host–gut microbiota interactions.
      • Sherriff J.L.
      • O'Sullivan T.A.
      • Properzi C.
      • et al.
      Choline, its potential role in nonalcoholic fatty liver disease, and the case for human and bacterial genes.
      The association between dietary choline deficiency and hepatic lipid accumulation has been recognized for more than 50 years
      • Nakamura T.
      • Nakamura S.
      • Karoji N.
      • et al.
      Hepatic function tests in heavy drinkers among workmen.
      and is routinely used to stimulate NAFLD in animal models. Human dietary choline requirements vary depending on estrogen status and genetic polymorphisms.
      • da Costa K.A.
      • Corbin K.D.
      • Niculescu M.D.
      • et al.
      Identification of new genetic polymorphisms that alter the dietary requirement for choline and vary in their distribution across ethnic and racial groups.
      From a cross-sectional analysis of patients in the NASH Clinical Research Network, deficiency of choline was associated with a worsening of liver fibrosis in postmenopausal women even after adjusting for common factors (age, obesity, T2D, serum triglycerides, steroid use) linked to NAFLD in multiple ordinal logistic regression models.
      • Guerrerio A.L.
      • Colvin R.M.
      • Schwartz A.K.
      • et al.
      Choline intake in a large cohort of patients with nonalcoholic fatty liver disease.
      A significant negative correlation between scores of fatty liver index (FLI) and choline consumption was recently reported in a study of 20,643 persons, with a 14% lower risk of NAFLD in those with the highest choline intake; those with higher BMI had greater reductions in FLI with increasing choline intake.
      • Mazidi M.
      • Katsiki N.
      • Mikhailidis D.P.
      • et al.
      Adiposity may moderate the link between choline intake and non-alcoholic fatty liver disease.
      Although associations between dietary choline and NAFLD exist, no controlled interventional studies are currently available; therefore, the precise role for choline/phosphatidylcholine supplementation in the progression of NAFLD to steatohepatitis and serious hepatic consequences remains to be elucidated.

      Polyphenols

      Polyphenols, commonly found in foods such as fruit, vegetables, wine, and coffee, have been shown to reduce IHTG through several mechanisms, including inhibition of lipogenesis via SREBP1c downregulation and by inducing antioxidant and anti-inflammatory effects.
      • Rodriguez-Ramiro I.
      • Vauzour D.
      • Minihane A.M.
      Polyphenols and non-alcoholic fatty liver disease: impact and mechanisms.
      Although polyphenol deficiency per se is poorly understood in patients with NAFLD, various members of the polyphenol family, such as resveratrol, curcumin, quercetin, and green tea catechins, have shown a potential to reduce lipid peroxidation, liver enzyme levels, and inflammatory biomarker levels.
      • Wu C.H.
      • Lin M.C.
      • Wang H.C.
      • et al.
      Rutin inhibits oleic acid induced lipid accumulation via reducing lipogenesis and oxidative stress in hepatocarcinoma cells.
      • Shimada T.
      • Tokuhara D.
      • Tsubata M.
      • et al.
      Flavangenol (pine bark extract) and its major component procyanidin B1 enhance fatty acid oxidation in fat-loaded models.
      • Chang H.C.
      • Peng C.H.
      • Yeh D.M.
      • et al.
      Hibiscus sabdariffa extract inhibits obesity and fat accumulation, and improves liver steatosis in humans.
      For example, daily supplementation with both 150 mg and 500 mg of resveratrol reduced total cholesterol and aspartate transaminase (AST)/ALT levels while improving both insulin sensitivity and NAFLD pathogenesis.
      • Faghihzadeh F.
      • Adibi P.
      • Rafiei R.
      • et al.
      Resveratrol supplementation improves inflammatory biomarkers in patients with nonalcoholic fatty liver disease.
      However, no clinical trials incorporating polyphenol supplementation have collected liver biopsies; therefore, histologic data are not currently available.

      Vitamin D

      Recent epidemiologic evidence shows that patients with NAFLD are more frequently deficient in vitamin D than the general population, and circulating vitamin D levels are proportional to the degree of fibrotic evolution.
      • Keane J.T.
      • Elangovan H.
      • Stokes R.A.
      • et al.
      Vitamin D and the liver-correlation or cause?.
      In cell and rodent models, vitamin D supplementation produced multiple beneficial effects, from improvements in insulin sensitivity to anti-inflammatory effects in both adipose and liver to slowing down hepatic fibrosis.
      • Mazzone G.
      • Morisco C.
      • Lembo V.
      • et al.
      Dietary supplementation of vitamin D prevents the development of western diet-induced metabolic, hepatic and cardiovascular abnormalities in rats.
      However, despite 24 weeks of 2000 IU/d vitamin D supplementation in 65 patients with T2D with NAFLD, neither hepatic steatosis nor markers of inflammation, fibrosis, or cardiovascular/metabolic parameters improved.
      • Barchetta I.
      • Del Ben M.
      • Angelico F.
      • et al.
      No effects of oral vitamin D supplementation on non-alcoholic fatty liver disease in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled trial.
      In a more recent study of 2423 prediabetic adults, Pittas and colleagues
      • Pittas A.G.
      • Dawson-Hughes B.
      • Sheehan P.
      • et al.
      Vitamin D supplementation and prevention of type 2 diabetes.
      showed that, despite 24 months of 4000-IU/d vitamin D supplementation, which doubled serum vitamin D levels, risk of new-onset diabetes was not significantly decreased compared with placebo.

      Vitamin E

      In addition to being one of the most potent antioxidants in nature, vitamin E is regarded as the main lipid-soluble antioxidant involved in the regulation of gene expression, inflammatory responses, and modulation of cellular signaling.
      • Perumpail B.J.
      • Li A.A.
      • John N.
      • et al.
      The role of vitamin E in the treatment of NAFLD.
      Sources of vitamin E are similar to those of polyunsaturated FAs and include olive oil, nuts, and green vegetables; therefore, vitamin E deficiency is also likely to be observed in the typical Western diet. Vitamin E supplementation, prescribed as either monotherapy or as part of a combination therapy approach, has successfully improved liver histology scores and reduced the odds of hepatic steatosis in patients with NAFLD and NASH.
      • El Hadi H.
      • Vettor R.
      • Rossato M.
      Vitamin E as a treatment for nonalcoholic fatty liver disease: reality or myth?.
      Findings from the landmark PIVENS (Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis) clinical trial showed vitamin E supplementation had greater reductions in hepatocyte ballooning and lobular inflammation compared with pioglitazone treatment.
      • Sanyal A.J.
      • Chalasani N.
      • Kowdley K.V.
      • et al.
      Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis.
      Consequently, both the American Association for the Study of Liver Diseases and the European Association for the Study of the Liver guidelines recognize vitamin E supplementation as an effective short-term treatment option for nondiabetic patients with biopsy-proven NASH.
      European Association for the Study of the LiverEuropean Association for the Study of DiabetesEuropean Association for the Study of Obesity
      EASL-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease.

      Iron, copper, zinc

      Although an essential nutrient in multiple cellular processes and erythropoiesis, an excessive amount of iron is commonly observed in patients with NAFLD and is associated with organ dysfunction secondary to the formation of reactive oxygen species.
      • Dongiovanni P.
      • Lanti C.
      • Gatti S.
      • et al.
      High fat diet subverts hepatocellular iron uptake determining dysmetabolic iron overload.
      Furthermore, subversion of iron metabolism and an increased facilitation of iron storage have been reported following hepatocyte exposure to FFAs and in patients with NAFLD and copper deficiency.
      • Aigner E.
      • Theurl I.
      • Haufe H.
      • et al.
      Copper availability contributes to iron perturbations in human nonalcoholic fatty liver disease.
      Copper deficiency is observed in human NAFLD and is associated with insulin resistance, steatosis, and an accelerated progression of NASH.
      • Aigner E.
      • Strasser M.
      • Haufe H.
      • et al.
      A role for low hepatic copper concentrations in nonalcoholic Fatty liver disease.
      Moreover, dietary copper deficiency and fructose feeding synergistically exacerbate liver damage and accelerate hepatic fat accumulation, inflammation, and fibrogenesis.
      • Fields M.
      • Holbrook J.
      • Scholfield D.
      • et al.
      Effect of fructose or starch on copper-67 absorption and excretion by the rat.
      ,
      • Song M.
      • Schuschke D.A.
      • Zhou Z.
      • et al.
      High fructose feeding induces copper deficiency in Sprague-Dawley rats: a novel mechanism for obesity related fatty liver.
      The association between liver disease and zinc deficiency has been recognized for more than half a century.
      • Mohammad M.K.
      • Zhou Z.
      • Cave M.
      • et al.
      Zinc and liver disease.
      Zinc deficiency initiates insulin resistance, iron overload, and hepatic steatosis, which follows the impairment of zinc homeostasis caused by chronic liver disease.
      • Himoto T.
      • Masaki T.
      Associations between zinc deficiency and metabolic abnormalities in patients with chronic liver disease.
      Taken together, the ready accessibility of energy-dense foods, market saturation of hidden sugars in foods such as breakfast cereals, and a general trend toward highly processed convenience foods contribute to an imbalanced, nutrient-poor diet and, consequently, metabolic disease in Western society.
      • Hruby A.
      • Hu F.B.
      The epidemiology of obesity: a big picture.
      As described earlier, the imbalance and inadequacies of specific macronutrients and micronutrients affects key processes associated with NAFLD pathogenesis and progression (see Figs. 1 and 2). These observations also highlight another key concept that is further expanded below: it is the ratio and the type of nutrients, in addition to the absolute amount or caloric value, that dictates long-term health.
      • Solon-Biet S.M.
      • McMahon A.C.
      • Ballard J.W.
      • et al.
      The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice.

      Modulation of metabolism: interventions for nonalcoholic fatty liver disease with diet and physical activity

      Regulation of energy balance is highly complex and dynamically interrelated such that a simplistic arithmetical approach of calories in and calories out does not account for counterbalancing homeostatic processes and is unsubstantiated by current evidence.
      • Hall K.D.
      • Guo J.
      Obesity energetics: body weight regulation and the effects of diet composition.
      Thus, the authors advocate modulating metabolism in an integrated manner to simultaneously affect the interdependent variables of energy intake and expenditure (Fig. 3).
      Figure thumbnail gr3
      Fig. 3Modulation of energy metabolism in NAFLD treatment. Simultaneous modulation of energy output and energy intake can be used as part of a comprehensive NAFLD treatment regimen. Implementing PA as well as consuming diets that are rich in bioactive nutrients such as FAs, amino acids, and polyphenols can contribute to improvements in NAFLD and related parameters. Dietary changes that encompass restriction of certain nutrients (eg, CHO) or balanced nutrition via the Mediterranean diet could also be used in the treatment of NAFLD. Currently available data support the Mediterranean diet as a consistently effective dietary approach to produce benefits on metabolic syndrome, NAFLD, T2D, and CVD. BHB, β-hydroxybutyrate; CHOL, cholesterol; DAG, diacylglycerol.

      Metabolic Modulation by Dietary Changes: Restriction, Induction, Balance

      The individual macronutrients and micronutrients discussed earlier have shown the ability to improve measures of liver and metabolic health through various biological mechanisms. It follows that adjusting the composition of the diet by consumption of foods with bioactive components, such as specific FAs, amino acids, and polyphenols, should be considered as part of a comprehensive dietary approach for NAFLD. At present, the most consistent and robust evidence seems to be for a balanced diet versus nutrient restriction or induction, all of which are briefly reviewed here.

      Carbohydrate restriction

      CHO restriction as a successful strategy to induce weight loss may be partly explained via suppression of the usual weight loss–mediated increase in ghrelin level.
      • Sumithran P.
      • Prendergast L.A.
      • Delbridge E.
      • et al.
      Ketosis and appetite-mediating nutrients and hormones after weight loss.
      In addition, CHO restriction is of significant clinical interest because its proposed metabolic benefits do not rely on achieving weight loss.
      • Cox P.J.
      • Clarke K.
      Acute nutritional ketosis: implications for exercise performance and metabolism.
      However, research on CHO restriction is often confounded by weight loss and protein/fat composition of the CHO-restricted interventional diet.
      For example, a short, 14-day intervention with an isocaloric, CHO-restricted, increased-protein diet in 17 obese individuals with NAFLD reduced liver fat levels paralleled by (1) decreases in hepatic de novo lipogenesis, (2) increases in serum β-hydroxybutyrate concentration, (3) increases in folate-producing Streptococcus and serum folate concentration, and (4) downregulation of the lipogenic pathway with upregulation of folate-mediated 1-carbon metabolism and FA oxidation pathways from transcriptomic analysis of liver biopsy samples (see Fig. 3).
      • Mardinoglu A.
      • Wu H.
      • Bjornson E.
      • et al.
      An integrated understanding of the rapid metabolic benefits of a carbohydrate-restricted diet on hepatic steatosis in humans.
      However, the protein content within this interventional CHO-restricted diet was increased by 33% from the baseline diet.
      • Mardinoglu A.
      • Wu H.
      • Bjornson E.
      • et al.
      An integrated understanding of the rapid metabolic benefits of a carbohydrate-restricted diet on hepatic steatosis in humans.
      Other CHO-restricted studies in which the CHO was replaced almost entirely with dietary fat had either no reduction or showed an increase in IHTG levels.
      • Haufe S.
      • Engeli S.
      • Kast P.
      • et al.
      Randomized comparison of reduced fat and reduced carbohydrate hypocaloric diets on intrahepatic fat in overweight and obese human subjects.
      A further complicating factor is whether the fat comprises predominantly unsaturated or saturated fats.
      • Westerbacka J.
      • Lammi K.
      • Hakkinen A.M.
      • et al.
      Dietary fat content modifies liver fat in overweight nondiabetic subjects.
      Thus, the role of the replacing macronutrient on liver fat in the context of CHO restriction needs to be further clarified.

      Induction of ketogenesis

      Insufficient ketogenesis caused by mitochondrial dysfunction is a key pathogenic defect in the NAFL to NASH transition.
      • Sunny N.E.
      • Parks E.J.
      • Browning J.D.
      • et al.
      Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease.
      Effects of ketogenic diets (KDs) in both rodents and humans with NAFLD remain unclear. In rodent models, KD feeding resulted in hepatic insulin resistance, increased hepatic diacylglycerol content, and increased lipid accumulation and inflammation (see Fig. 3).
      • Jornayvaz F.R.
      • Jurczak M.J.
      • Lee H.Y.
      • et al.
      A high-fat, ketogenic diet causes hepatic insulin resistance in mice, despite increasing energy expenditure and preventing weight gain.
      ,
      • Ellenbroek J.H.
      • van Dijck L.
      • Tons H.A.
      • et al.
      Long-term ketogenic diet causes glucose intolerance and reduced beta- and alpha-cell mass but no weight loss in mice.
      Hepatic insulin resistance was induced with both short-term and long-term KD feeding, despite increased energy expenditure and weight gain prevention.
      • Jornayvaz F.R.
      • Jurczak M.J.
      • Lee H.Y.
      • et al.
      A high-fat, ketogenic diet causes hepatic insulin resistance in mice, despite increasing energy expenditure and preventing weight gain.
      ,
      • Ellenbroek J.H.
      • van Dijck L.
      • Tons H.A.
      • et al.
      Long-term ketogenic diet causes glucose intolerance and reduced beta- and alpha-cell mass but no weight loss in mice.
      In humans, effects of nutritional ketosis improved liver fat and NAFLD fibrosis score
      • Vilar-Gomez E.
      • Athinarayanan S.J.
      • Adams R.N.
      • et al.
      Post hoc analyses of surrogate markers of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis in patients with type 2 diabetes in a digitally supported continuous care intervention: an open-label, non-randomised controlled study.
      while decreased fasting plasma insulin, triglycerides, and FGF21 levels.
      • Rosenbaum M.
      • Hall K.D.
      • Guo J.
      • et al.
      Glucose and lipid homeostasis and inflammation in humans following an isocaloric ketogenic diet.
      However, these studies are significantly confounded by weight loss
      • Vilar-Gomez E.
      • Athinarayanan S.J.
      • Adams R.N.
      • et al.
      Post hoc analyses of surrogate markers of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis in patients with type 2 diabetes in a digitally supported continuous care intervention: an open-label, non-randomised controlled study.
      and CHO restriction,
      • Rosenbaum M.
      • Hall K.D.
      • Guo J.
      • et al.
      Glucose and lipid homeostasis and inflammation in humans following an isocaloric ketogenic diet.
      making it difficult to ascertain whether the beneficial effects are driven directly by the increase in plasma ketone levels. Ketosis induced by CHO restriction was associated with increased levels of cholesterol and inflammatory markers (see Fig. 3) and decreased insulin-mediated antilipolysis in overweight and obese men.
      • Rosenbaum M.
      • Hall K.D.
      • Guo J.
      • et al.
      Glucose and lipid homeostasis and inflammation in humans following an isocaloric ketogenic diet.
      To circumvent the unwanted dietary restriction of adhering to a caloric-restricted or CHO-restricted diet, or a KD, and therefore to examine a pure ketone-driven effect, an edible form of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate ketone ester (KE) was administered to healthy human participants.
      • Cox P.J.
      • Kirk T.
      • Ashmore T.
      • et al.
      Nutritional ketosis alters fuel preference and thereby endurance performance in athletes.
      At rest, KE intake induced a 3-fold increase in intramuscular concentrations of β-hydroxybutyrate and suppression of all measured muscle glycolytic intermediates after an exercise bout. This physiologic state induced by exogenous KE is opposite to that of endogenous ketosis induced by low CHO intake, in which replete glucose reserves, an intact insulin axis, and increased levels of ketone bodies would never usually coexist.
      • Cox P.J.
      • Kirk T.
      • Ashmore T.
      • et al.
      Nutritional ketosis alters fuel preference and thereby endurance performance in athletes.
      Whether these KEs also affect liver fat and other aspects of NAFLD pathogenesis remains to be determined.
      Because nutritional ketosis by isocaloric CHO restriction induces beta oxidation in both muscle
      • Cox P.J.
      • Kirk T.
      • Ashmore T.
      • et al.
      Nutritional ketosis alters fuel preference and thereby endurance performance in athletes.
      and liver,
      • Browning J.D.
      • Weis B.
      • Davis J.
      • et al.
      Alterations in hepatic glucose and energy metabolism as a result of calorie and carbohydrate restriction.
      it is surmised that such increase in fat oxidation would lead to increased energy expenditure and consequently to body fat loss, the basis of the original Atkins diet.
      • Atkins R.C.
      Atkins’ diet revolution: the high calorie way to stay thin forever.
      However, the experimental evidence does not support such a metabolic advantage. A systematic review and meta-analysis of 32 controlled-feeding studies (n = 563) by Hall and Guo
      • Hall K.D.
      • Guo J.
      Obesity energetics: body weight regulation and the effects of diet composition.
      on the effects on daily energy expenditure and body fat of isocaloric diets differing in their CHO and fat content but with equal protein found that both energy expenditure (26 kcal/d) and fat loss (16 g/d) were significantly greater with lower-fat diets. It is possible that KDs with the fat predominantly derived from unsaturated fat sources might produce reductions in liver fat. Nonetheless, in light of the available evidence, higher-protein diets seem to confer advantages on energy expenditure, body composition, and liver fat more than a CHO-restricted KD in humans.

      Balanced composition

      A consistently successful example of a dietary intervention to improve metabolic health is the Mediterranean diet (MEDd).
      • Esposito K.
      • Maiorino M.I.
      • Bellastella G.
      • et al.
      A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses.
      • Kastorini C.M.
      • Milionis H.J.
      • Esposito K.
      • et al.
      The effect of Mediterranean diet on metabolic syndrome and its components: a meta-analysis of 50 studies and 534,906 individuals.
      • Sofi F.
      • Casini A.
      Mediterranean diet and non-alcoholic fatty liver disease: new therapeutic option around the corner?.
      • Tong T.Y.
      • Wareham N.J.
      • Khaw K.T.
      • et al.
      Prospective association of the Mediterranean diet with cardiovascular disease incidence and mortality and its population impact in a non-Mediterranean population: the EPIC-Norfolk study.
      The MEDd is typically low in simple sugars and high in fiber, fresh fruit and vegetables, olives, avocados, nuts, and fish. These foods are rich in bioactive FAs, such as MUFAs and polyunsaturated FAs, and potent antioxidants, such as vitamin E and polyphenols, which are critical regulators of nutrient-sensed transcription factors (eg, SREBP-1c, PPARα, and PPARγ), which regulate core metabolic pathways (see Fig. 3).
      • Jump D.B.
      N-3 polyunsaturated fatty acid regulation of hepatic gene transcription.
      MEDd has been extensively studied in large multicenter trials and proved to decrease the incidence of the metabolic syndrome,
      • Kastorini C.M.
      • Milionis H.J.
      • Esposito K.
      • et al.
      The effect of Mediterranean diet on metabolic syndrome and its components: a meta-analysis of 50 studies and 534,906 individuals.
      T2D,
      • Esposito K.
      • Maiorino M.I.
      • Bellastella G.
      • et al.
      A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses.
      CVD,
      • Tong T.Y.
      • Wareham N.J.
      • Khaw K.T.
      • et al.
      Prospective association of the Mediterranean diet with cardiovascular disease incidence and mortality and its population impact in a non-Mediterranean population: the EPIC-Norfolk study.
      and NAFLD.
      • Sofi F.
      • Casini A.
      Mediterranean diet and non-alcoholic fatty liver disease: new therapeutic option around the corner?.
      In a randomized controlled, 6-week crossover dietary intervention study comparing a MEDd with low-fat/high-CHO diet in 12 insulin-resistant subjects with biopsy-proven NAFLD, MEDd significantly reduced mean IHTG levels and HOMA-IR by 39% and 36%, respectively, without change in body weight.
      • Ryan M.C.
      • Itsiopoulos C.
      • Thodis T.
      • et al.
      The Mediterranean diet improves hepatic steatosis and insulin sensitivity in individuals with non-alcoholic fatty liver disease.
      A study by Trovato and colleagues
      • Trovato F.M.
      • Martines G.F.
      • Brischetto D.
      • et al.
      Neglected features of lifestyle: their relevance in non-alcoholic fatty liver disease.
      (n = 1199) revealed that the Adherence to MEDd Score was a significant independent predictor of hepatic steatosis severity.
      A precise understanding of which specific components of the MEDd mediate the beneficial effects remains unclear. It most likely is a combination of all the nutrients discussed earlier. It is also difficult to precisely define a MEDd because it is a broad pattern. Nonetheless, the PREDIMED (Prevención con Dieta Mediterránea)
      • Estruch R.
      • Ros E.
      • Salas-Salvado J.
      • et al.
      Primary prevention of cardiovascular disease with a Mediterranean diet.
      study provided robust clinical trial evidence for intervention diets consisting of 40% CHO, 16% protein, and 44% fat, the fat being predominantly MUFAs (22% of calories) with saturated FAs at 9% and polyunsaturated FAs at 6% in the prevention of CVD. Thus, the authors propose a MEDd that replaces some CHO with protein (ideally 25%–30% calories from protein) as an optimal diet for NAFLD.

      Metabolic Modulation by Decreasing Energy Intake

      Irrespective of nutrient composition, both progressive (1200–1500 kcal/d restriction) and severe methods of calorie restriction, such as bariatric surgery, are effective in inducing weight loss and improving metabolic health outcomes in patients with NAFLD
      • McCarthy E.M.
      • Rinella M.E.
      The role of diet and nutrient composition in nonalcoholic Fatty liver disease.
      ,
      • Bellentani S.
      • Dalle Grave R.
      • Suppini A.
      • et al.
      Fatty Liver Italian N. Behavior therapy for nonalcoholic fatty liver disease: The need for a multidisciplinary approach.
      (see Fig. 3). For example, results from the Comprehensive Assessment of Long-Term Effects of Reducing Intake of Energy (CALERIE) study (n = 218) showed that 6 months of 25% calorie restriction while maintaining sufficient nutrition resulted in ∼10% progressive decline in body weight accompanied by 29% decrease in fasting plasma insulin level, 27% reduction in both subcutaneous and visceral fat, and 37% decrease in hepatic lipid level.
      • Redman L.M.
      • Ravussin E.
      Caloric restriction in humans: impact on physiological, psychological, and behavioral outcomes.
      Significant weight loss following bariatric surgery resulted in complete regression of NAFLD and necroinflammatory activity in 82% and 93%, respectively, in a study of 284 obese patients.
      • Weiner R.A.
      Surgical treatment of non-alcoholic steatohepatitis and non-alcoholic fatty liver disease.
      In a separate cohort of 109 morbidly obese patients with biopsy-proven NASH undergoing bariatric surgery, 85% had complete resolution of NASH, and fibrosis was reduced in 33.8% of patients after 1 year of follow-up.
      • Lassailly G.
      • Caiazzo R.
      • Buob D.
      • et al.
      Bariatric surgery reduces features of nonalcoholic steatohepatitis in morbidly obese patients.
      Another approach to decreasing energy input is by intermittent fasting (see Fig. 3), which has been shown to significantly reduce obesity, fasting insulin level, and leptin level.
      • Heilbronn L.K.
      • Smith S.R.
      • Martin C.K.
      • et al.
      Alternate-day fasting in nonobese subjects: effects on body weight, body composition, and energy metabolism.
      Systematic reviews of various fasting regimens seem to promote weight loss and suggest improvement in metabolic health.
      • Horne B.D.
      • Muhlestein J.B.
      • Anderson J.L.
      Health effects of intermittent fasting: hormesis or harm? A systematic review.
      • Patterson R.E.
      • Sears D.D.
      Metabolic effects of intermittent fasting.
      • Harris L.
      • Hamilton S.
      • Azevedo L.B.
      • et al.
      Intermittent fasting interventions for treatment of overweight and obesity in adults: a systematic review and meta-analysis.
      However, most studies to date have focused on changes related to body weight and in metabolic parameters for T2D and CVD, and not specifically for NAFLD. In addition, human studies have been limited to observational studies of religious fasting (eg, during Ramadan), cross-sectional studies of eating patterns, and small experimental studies, underscoring the need for robust study designs to adequately examine intermittent fasting in humans.
      Although calorie-reducing interventions could be effective short-term strategies to improve metabolic health, long-term adherence has not been sufficiently explored. It is likely that poor adherence to such approaches and regression to normal caloric consumption may limit the effectiveness, as shown by the high dropout rates
      • Trepanowski J.F.
      • Kroeger C.M.
      • Barnosky A.
      • et al.
      Effect of alternate-day fasting on weight loss, weight maintenance, and cardioprotection among metabolically healthy obese adults: a randomized clinical trial.
      and the inability to suppress hunger
      • Heilbronn L.K.
      • Smith S.R.
      • Martin C.K.
      • et al.
      Alternate-day fasting in nonobese subjects: effects on body weight, body composition, and energy metabolism.
      during alternate-day fasting interventions. Thus, education on optimal nutrient compositions should continue to be an integral consideration in the overall management of NAFLD.

      Metabolic Modulation by Increasing Energy Output

      Only a small reduction in body weight is necessary to elicit significant improvements in liver health and metabolic parameters. For example, a 25% to 43% reduction in IHTG levels has been reported following body weight loss of just 5%, with improvements seen in necroinflammation, reductions in NAFLD activity score, resolution of NASH, and regression of fibrosis with progressive weight loss of greater than 10%.
      • Patel N.S.
      • Doycheva I.
      • Peterson M.R.
      • et al.
      Effect of weight loss on magnetic resonance imaging estimation of liver fat and volume in patients with nonalcoholic steatohepatitis.
      • Magkos F.
      • Fraterrigo G.
      • Yoshino J.
      • et al.
      Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity.
      • Vilar-Gomez E.
      • Martinez-Perez Y.
      • Calzadilla-Bertot L.
      • et al.
      Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis.
      Although it is well established that both intake (decreased) and output (increased) must be modulated to attain net energy loss, the laws of thermodynamics make it highly inefficient to lose body weight purely via increasing energy output alone. Nonetheless, the literature strongly supports the role of PA in the management of NAFLD (see Fig. 3).
      • Marchesini G.
      • Petta S.
      • Dalle Grave R.
      Diet, weight loss, and liver health in nonalcoholic fatty liver disease: pathophysiology, evidence, and practice.
      ,
      • Johnson N.A.
      • Keating S.E.
      • George J.
      Exercise and the liver: implications for therapy in fatty liver disorders.
      ,
      • Oh S.
      • Shida T.
      • Yamagishi K.
      • et al.
      Moderate to vigorous physical activity volume is an important factor for managing nonalcoholic fatty liver disease: a retrospective study.
      In addition to improving body composition, PA also improves both biopsy-confirmed
      • Eckard C.
      • Cole R.
      • Lockwood J.
      • et al.
      Prospective histopathologic evaluation of lifestyle modification in nonalcoholic fatty liver disease: a randomized trial.
      ,
      • Promrat K.
      • Kleiner D.E.
      • Niemeier H.M.
      • et al.
      Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis.
      and noninvasive measures of steatosis and liver health independent of significant weight loss.
      • Golabi P.
      • Locklear C.T.
      • Austin P.
      • et al.
      Effectiveness of exercise in hepatic fat mobilization in non-alcoholic fatty liver disease: systematic review.
      • Bacchi E.
      • Negri C.
      • Targher G.
      • et al.
      Both resistance training and aerobic training reduce hepatic fat content in type 2 diabetic subjects with nonalcoholic fatty liver disease (the RAED2 Randomized Trial).
      • Slentz C.A.
      • Bateman L.A.
      • Willis L.H.
      • et al.
      Effects of aerobic vs. resistance training on visceral and liver fat stores, liver enzymes, and insulin resistance by HOMA in overweight adults from STRRIDE AT/RT.
      Literature pertaining to the optimal mode and intensity of PA for the management of NAFLD is conflicting.
      • Keating S.E.
      • Hackett D.A.
      • Parker H.M.
      • et al.
      Effect of aerobic exercise training dose on liver fat and visceral adiposity.
      ,
      • Kistler K.D.
      • Brunt E.M.
      • Clark J.M.
      • et al.
      Physical activity recommendations, exercise intensity, and histological severity of nonalcoholic fatty liver disease.
      In a prospective cohort study of 1149 people with baseline NAFL studied over 7 to 9 years, only vigorous-intensity PA (≥7 metabolic equivalent of tasks (MET)), but not moderate-low–intensity (3–5 MET) or moderate-high–intensity (5–7 MET) PA, was able to prevent the progression from NAFL to NASH.
      • Tsunoda K.
      • Kai Y.
      • Kitano N.
      • et al.
      Impact of physical activity on nonalcoholic steatohepatitis in people with nonalcoholic simple fatty liver: a prospective cohort study.
      Keating and colleagues
      • Keating S.E.
      • George J.
      • Johnson N.A.
      The benefits of exercise for patients with non-alcoholic fatty liver disease.
      advocate a prescription of 150 to 300 min/wk of moderate-intensity to vigorous-intensity (50%–70% Vo2 [maximum oxygen uptake] peak) aerobic exercise, performed on a minimum of 3 d/wk, which resulted in mean relative reduction in IHTG level of 28% for hepatic benefits in patients with NAFLD.
      The mechanisms mediating PA and improvements in metabolic disease are complex and still not fully understood. However, strong similarities between existing pharmacologic target approaches and PA on cellular metabolism have been identified. For example, both metformin and PA are potent activators of AMPK within skeletal muscle, adipose, and liver tissue that increase mitochondrial oxidative capacity and shift substrate metabolism toward fat oxidation while downregulating lipogenic enzymes by suppressing SREBP-1c.
      • Richter E.A.
      • Ruderman N.B.
      AMPK and the biochemistry of exercise: implications for human health and disease.
      PA is also a potent stimulator of PPARα, which activates beta oxidation,
      • Wu H.
      • Jin M.
      • Han D.
      • et al.
      Protective effects of aerobic swimming training on high-fat diet induced nonalcoholic fatty liver disease: regulation of lipid metabolism via PANDER-AKT pathway.
      ,
      • Rector R.S.
      • Thyfault J.P.
      • Morris R.T.
      • et al.
      Daily exercise increases hepatic fatty acid oxidation and prevents steatosis in Otsuka Long-Evans Tokushima Fatty rats.
      improves oxidative stress and innate immune system activation,
      • Richter E.A.
      • Ruderman N.B.
      AMPK and the biochemistry of exercise: implications for human health and disease.
      and increases glucose transporter type 4 expression, which facilitates glucose disposal within skeletal muscles to enhance insulin sensitivity.
      • Richter E.A.
      • Hargreaves M.
      Exercise, GLUT4, and skeletal muscle glucose uptake.
      In aggregate, taking the best evidence to date and, in particular, the powerful effects of certain nutrients in food on human physiology, the authors recommend a dietary framework for NAFLD as shown in Table 2. This construct could serve as a standardizing approach to dietary recommendations in a clinical practice setting, as well as provide a foundation for dietary control in NAFLD clinical trials given the substantial placebo effect in this disease setting.
      Table 2Dietary recommendations for nonalcoholic fatty liver disease based on the available current evidence
      • Prioritize intact starches such as brown rice, quinoa, and steel-cut oats, and limit or avoid refined starches such as white bread and white rice
      • Replace some of the CHO, especially refined CHO, in the diet with additional protein from a mixture of animal or vegetable sources, including chicken, fish, cheese, tofu, and pulses
      • Include a variety of bioactive compounds in the diet by consuming fruits, vegetables, coffee, tea, nuts, seeds, and extra virgin olive oil
      • Get most fat from unsaturated sources, such as olive oil (ideally extra virgin), rapeseed oil, sunflower oil, safflower oil, canola oil, or nuts and seeds
      • Limit or avoid added sugars, whether sucrose, fructose, maltose, maltodextrin, or any syrups. If any of these words appear in the first 3–5 ingredients of any food item, it is best to avoid that item and choose a no-sugar version instead. Examples are yogurts and commercial cereals
      • In particular, avoid liquid sugar such as carbonated sugary drinks/sodas, lemonade, any juices, smoothies, and added sugar to tea and coffee
      Typical Daily Menu
      Exact portion sizes differ based on an individual's energy requirements. Energy balance is the biggest driver of liver fat change; therefore, a weight-maintaining or weight-reducing diet should be advised if an individual is overweight.
      • Breakfast
        • Oatmeal
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
          ,
          Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
          made with:
          • steel-cut oats
          • Semiskimmed milk,
            Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
            blueberrries,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            raspberries,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            sliced almonds
            Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
            ,
            Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
            ,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            ,
            Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
            ,
            Although foods rich in unsaturated fats rather than saturated fats should be emphasized, there is some evidence that fermented dairy foods, including yogurts, have a neutral or even protective effect against cardiovascular disease.
        • Black coffee
          Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
      • Lunch
        • Smoked salmon
          Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
          ,
          Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
          salad made with:
          • Rocket (arugula),
            Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
            chicory greens,
            Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
            ,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            tomatoes,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            cucumber,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            extravirgin olive oil,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            ,
            Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
            and pine nuts
            Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
            ,
            Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
            ,
            Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
            ,
            Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
        • Apple
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
      • Dinner
        • Chicken breast
          Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
        • Tabbouleh
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
        • Sliced avocado
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
          ,
          Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
        • Hummus
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
          ,
          Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
          ,
          Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
        • Chopped vegetables
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
          ,
          Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
        • Whole yogurt
          Although foods rich in unsaturated fats rather than saturated fats should be emphasized, there is some evidence that fermented dairy foods, including yogurts, have a neutral or even protective effect against cardiovascular disease.
          with pecans
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
          ,
          Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
          ,
          Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
          ,
          Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
          and blackberries
          Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
          ,
          Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
      a Exact portion sizes differ based on an individual's energy requirements. Energy balance is the biggest driver of liver fat change; therefore, a weight-maintaining or weight-reducing diet should be advised if an individual is overweight.
      b Partly or completely indigestible CHO increase the production of SCFAs via colon fermentation and gut microbiota, which modulate whole-body insulin sensitivity via a variety of mechanisms, as discussed in this article. The low-glycemic load of this diet has also been linked with lower liver fat levels.
      c Proteins and amino acids could decrease IHTG levels without weight loss via several mechanisms that affect metabolism, inflammation, oxidative stress, and gut epithelial barrier physiology.
      d Foods rich in polyphenols and other bioactive compounds may decrease inflammatory pathways implicated in liver disease.
      e Unsaturated fats, such as n-3 or n-6 polyunsaturated FAs, or MUFAs, have been shown to improve insulin sensitivity and decrease IHTG levels when they replace saturated fat.
      f Although foods rich in unsaturated fats rather than saturated fats should be emphasized, there is some evidence that fermented dairy foods, including yogurts, have a neutral or even protective effect against cardiovascular disease.

      Summary and future directions

      The concept of energy balance to allay metabolic maladies was recognized nearly half a millennium ago; however, it is only in the last few decades that clinicians have begun to systematically understand the biochemical and molecular underpinnings of those initial observations. Lifestyle-related factors remain the single biggest modifiable component of people’s health. The current evidence suggests that it is possible to reduce the burden of diseases related to caloric excess and disordered metabolism, such as NAFLD, with a structured approach to diet and PA. Experimental studies have started to identify mechanisms by which certain dietary nutrients may exert influence on the multifactorial aspects of NAFLD and provide a strong scientific foundation to support the possibility for combinatorial effects of nutritive components of food.
      Although current evidence strongly supports the role of lifestyle modification as a foundation for the management of NAFLD,
      • Chalasani N.
      • Younossi Z.
      • Lavine J.E.
      • et al.
      The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases.
      ,
      • Romero-Gomez M.
      • Zelber-Sagi S.
      • Trenell M.
      Treatment of NAFLD with diet, physical activity and exercise.
      it is also clear from the increasing incidence of NAFLD that such an approach alone may not be sufficient or sustainable, especially for more severe stages of the disease. Large changes (eg, in FA compositions) in the diet are often needed for modest (0.5%) absolute reductions in liver fat levels.
      • Bjermo H.
      • Iggman D.
      • Kullberg J.
      • et al.
      Effects of n-6 PUFAs compared with SFAs on liver fat, lipoproteins, and inflammation in abdominal obesity: a randomized controlled trial.
      The amounts of macronutrients and micronutrients differ depending on the food source and on the individual’s activity level or muscle mass, and optimal amounts of various foods still remain to be elucidated.
      • Elmadfa I.
      • Meyer A.L.
      Importance of food composition data to nutrition and public health.
      Although modest weight loss can produce significant effects on IHTG,
      • Patel N.S.
      • Doycheva I.
      • Peterson M.R.
      • et al.
      Effect of weight loss on magnetic resonance imaging estimation of liver fat and volume in patients with nonalcoholic steatohepatitis.
      • Magkos F.
      • Fraterrigo G.
      • Yoshino J.
      • et al.
      Effects of moderate and subsequent progressive weight loss on metabolic function and adipose tissue biology in humans with obesity.
      • Vilar-Gomez E.
      • Martinez-Perez Y.
      • Calzadilla-Bertot L.
      • et al.
      Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis.
      up to 50% of lost weight on average is regained by 1-year follow-up, with nearly all remaining lost weight regained thereafter in most individuals.
      • Butryn M.L.
      • Webb V.
      • Wadden T.A.
      Behavioral treatment of obesity.
      Thus, although dietary measures should remain integral to any NAFLD management approach, it is likely that additional interventions are also needed that are not solely dependent on weight loss to achieve benefits on NAFLD pathogenesis and progression.
      Recent advances in large-scale omics data-mining, machine learning, and systems biology are now allowing cross-disciplinary predictive insights to harness the power of people’s own bodies to fight disease and restore health, opening the possibility toward promising interventions beyond a dietary approach, such as leveraging key endogenous bioactive signaling intermediates, which may also exert influence on some of the same biological pathways and molecular targets as pharmacotherapies.
      • Dewar S.L.
      • Porter J.
      The effect of evidence-based nutrition clinical care pathways on nutrition outcomes in adult patients receiving non-surgical cancer treatment: a systematic review.
      Bespoke combinations of such endogenous metabolic modulators have the potential to be used as treatments in combination with both an underlying dietary framework as well as possible pharmacologic agents, expanding therapeutic options for patients across the NAFLD spectrum. Adequately controlled randomized clinical trials of sufficient duration and size are needed to determine whether such approaches are tractable in the real world and can form a mainstay of public health policy and recommendations for the management of NAFLD.

      Acknowledgments

      Editing assistance was provided by Caryne Craige, PhD, of Fishawack Communications Inc; funding for this assistance was provided by Axcella Health, Inc.

      Disclosures

      M.V. Chakravarthy is an employee of Axcella Health, Inc, and holds stock options in the company. T. Waddell and R. Banerjee are employees of Perspectum Diagnostics and hold shares and share options in the company. N. Guess has received research and fellowship funding from Diabetes UK, Diabetes Research and Wellness Foundation, Medical Research Council, Winston Churchill Memorial Trust, the American Overseas Dietetic Association, and Oviva. N. Guess has received consulting fees from Fixing Dad (a low-CHO app), Babylon Health, and Boehringer Ingelheim.
      This article reflects the independent perspectives of the individual authors and their interpretations of the scientific literature; it does not represent the views or positions of their respective employers/institutions.

      References

      1. Hippocrates. De Prisca Medicina. 400 BC.

        • Morgagni J.B.
        Epistola Anatoma Clinica XXI.
        New York Academy of Medicine, New York1765
        • Osler W.
        Lectures on angina pectoris and allied states.
        Appleton, New York1897
        • Nonni L.
        Diaeteticon, sive, De re cibaria libri IV.
        Antverpiae: Apud Petrum et Ioannem Belleros, 1627
        • Chalasani N.
        • Younossi Z.
        • Lavine J.E.
        • et al.
        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
        • Romero-Gomez M.
        • Zelber-Sagi S.
        • Trenell M.
        Treatment of NAFLD with diet, physical activity and exercise.
        J Hepatol. 2017; 67: 829-846
        • Dongiovanni P.
        • Valenti L.
        A nutrigenomic approach to non-alcoholic fatty liver disease.
        Int J Mol Sci. 2017; 18: E1534
        • Juarez-Hernandez E.
        • Chavez-Tapia N.C.
        • Uribe M.
        • et al.
        Role of bioactive fatty acids in nonalcoholic fatty liver disease.
        Nutr J. 2016; 15: 72
        • Marchesini G.
        • Petta S.
        • Dalle Grave R.
        Diet, weight loss, and liver health in nonalcoholic fatty liver disease: pathophysiology, evidence, and practice.
        Hepatology. 2016; 63: 2032-2043
        • McCarthy E.M.
        • Rinella M.E.
        The role of diet and nutrient composition in nonalcoholic Fatty liver disease.
        J Acad Nutr Diet. 2012; 112: 401-409
      2. World Health Organization. 2008 (Available at:) (Accessed August 20, 2019)
        • McKay A.
        • Wilman H.R.
        • Dennis A.
        • et al.
        Measurement of liver iron by magnetic resonance imaging in the UK Biobank population.
        PLoS One. 2018; 13: e0209340
        • Younossi Z.M.
        The epidemiology of nonalcoholic steatohepatitis.
        Clin Liver Dis (Hoboken). 2018; 11: 92-94
        • Dai W.
        • Ye L.
        • Liu A.
        • et al.
        Prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: a meta-analysis.
        Medicine (Baltimore). 2017; 96: e8179
        • Allen A.M.
        • Therneau T.M.
        • Larson J.J.
        • et al.
        Nonalcoholic fatty liver disease incidence and impact on metabolic burden and death: a 20 year-community study.
        Hepatology. 2018; 67: 1726-1736
        • Lobstein T.
        • Jackson-Leach R.
        Estimated burden of paediatric obesity and co-morbidities in Europe. Part 2. Numbers of children with indicators of obesity-related disease.
        Int J Pediatr Obes. 2006; 1: 33-41
        • Schwimmer J.B.
        • Deutsch R.
        • Kahen T.
        • et al.
        Prevalence of fatty liver in children and adolescents.
        Pediatrics. 2006; 118: 1388-1393
        • Public Health England
        Government dietary recommendations: government recommendations for energy and nutrients for males and females aged 1–18 years and 19+ years.
        Public Health England, London2016
        • Bennett E.
        • Peters S.A.E.
        • Woodward M.
        Sex differences in macronutrient intake and adherence to dietary recommendations: findings from the UK Biobank.
        BMJ Open. 2018; 8: e020017
        • Montague C.T.
        • Farooqi I.S.
        • Whitehead J.P.
        • et al.
        Congenital leptin deficiency is associated with severe early-onset obesity in humans.
        Nature. 1997; 387: 903-908
        • Wermter A.K.
        • Reichwald K.
        • Buch T.
        • et al.
        Mutation analysis of the MCHR1 gene in human obesity.
        Eur J Endocrinol. 2005; 152: 851-862
        • Bray G.A.
        • Dahms W.T.
        • Swerdloff R.S.
        • et al.
        The Prader-Willi syndrome: a study of 40 patients and a review of the literature.
        Medicine (Baltimore). 1983; 62: 59-80
        • Hruby A.
        • Hu F.B.
        The epidemiology of obesity: a big picture.
        Pharmacoeconomics. 2015; 33: 673-689
        • Cohen J.C.
        • Horton J.D.
        • Hobbs H.H.
        Human fatty liver disease: old questions and new insights.
        Science. 2011; 332: 1519-1523
        • Machado M.V.
        • Diehl A.M.
        Pathogenesis of nonalcoholic steatohepatitis.
        Gastroenterology. 2016; 150: 1769-1777
        • Galgani J.E.
        • Moro C.
        • Ravussin E.
        Metabolic flexibility and insulin resistance.
        Am J Physiol Endocrinol Metab. 2008; 295: E1009-E1017
        • Mota M.
        • Banini B.A.
        • Cazanave S.C.
        • et al.
        Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease.
        Metabolism. 2016; 65: 1049-1061
        • Angulo P.
        • Kleiner D.E.
        • Dam-Larsen S.
        • et al.
        Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease.
        Gastroenterology. 2015; 149: 389-397.e10
        • Ekstedt M.
        • Hagstrom H.
        • Nasr P.
        • et al.
        Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up.
        Hepatology. 2015; 61: 1547-1554
        • Bozaykut P.
        • Sahin A.
        • Karademir B.
        • et al.
        Endoplasmic reticulum stress related molecular mechanisms in nonalcoholic steatohepatitis.
        Mech Ageing Dev. 2016; 157: 17-29
        • Kanda T.
        • Matsuoka S.
        • Yamazaki M.
        • et al.
        Apoptosis and non-alcoholic fatty liver diseases.
        World J Gastroenterol. 2018; 24: 2661-2672
        • Mouries J.
        • Brescia P.
        • Silvestri A.
        • et al.
        Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development.
        J Hepatol. 2019; https://doi.org/10.1016/j.jhep.2019.08.005
        • Younossi Z.
        • Ratziu V.
        • Loomba R.
        • et al.
        Positive results from REGENERATE: a phase 3 international, randomized, placebo-controlled study evaluating obeticholic acid treatment for NASH.
        J Hepatol. 2019; 70: e5
        • Patel P.J.
        • Hayward K.L.
        • Rudra R.
        • et al.
        Multimorbidity and polypharmacy in diabetic patients with NAFLD: implications for disease severity and management.
        Medicine (Baltimore). 2017; 96: e6761
        • De Angelis M.
        • Garruti G.
        • Minervini F.
        • et al.
        The food-gut human axis: the effects of diet on gut microbiota and metabolome.
        Curr Med Chem. 2017; https://doi.org/10.2174/0929867324666170428103848
        • Scalbert A.
        • Brennan L.
        • Manach C.
        • et al.
        The food metabolome: a window over dietary exposure.
        Am J Clin Nutr. 2014; 99: 1286-1308
        • Esposito K.
        • Maiorino M.I.
        • Bellastella G.
        • et al.
        A journey into a Mediterranean diet and type 2 diabetes: a systematic review with meta-analyses.
        BMJ Open. 2015; 5: e008222
        • Kastorini C.M.
        • Milionis H.J.
        • Esposito K.
        • et al.
        The effect of Mediterranean diet on metabolic syndrome and its components: a meta-analysis of 50 studies and 534,906 individuals.
        J Am Coll Cardiol. 2011; 57: 1299-1313
        • Sofi F.
        • Casini A.
        Mediterranean diet and non-alcoholic fatty liver disease: new therapeutic option around the corner?.
        World J Gastroenterol. 2014; 20: 7339-7346
        • Tong T.Y.
        • Wareham N.J.
        • Khaw K.T.
        • et al.
        Prospective association of the Mediterranean diet with cardiovascular disease incidence and mortality and its population impact in a non-Mediterranean population: the EPIC-Norfolk study.
        BMC Med. 2016; 14: 135
        • Tilman D.
        • Clark M.
        Global diets link environmental sustainability and human health.
        Nature. 2014; 515: 518-522
        • Kotecha R.
        • Takami A.
        • Espinoza J.L.
        Dietary phytochemicals and cancer chemoprevention: a review of the clinical evidence.
        Oncotarget. 2016; 7: 52517-52529
        • Dewar S.L.
        • Porter J.