Elsevier

Metabolism

Volume 65, Issue 8, August 2016, Pages 1038-1048
Metabolism

Nonalcoholic Fatty Liver Disease: From Pathogenesis to Emerging Treatment
The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD)

https://doi.org/10.1016/j.metabol.2015.12.012Get rights and content

Abstract

Nonalcoholic fatty liver disease (NAFLD) is increasingly prevalent and represents a growing challenge in terms of prevention and treatment. Despite its high prevalence, only a small minority of affected patients develops inflammation and subsequently fibrosis and chronic liver disease, while most of them only exhibit simple steatosis. In this context, the full understanding of the mechanisms underlying the development of NAFLD and non-alcoholic steatohepatitis (NASH) is of extreme importance; despite advances in this field, knowledge on the pathogenesis of NAFLD is still incomplete. The ‘two-hit’ hypothesis is now obsolete, as it is inadequate to explain the several molecular and metabolic changes that take place in NAFLD. The “multiple hit” hypothesis considers multiple insults acting together on genetically predisposed subjects to induce NAFLD and provides a more accurate explanation of NAFLD pathogenesis. Such hits include insulin resistance, hormones secreted from the adipose tissue, nutritional factors, gut microbiota and genetic and epigenetic factors. In this article, we review the factors that form this hypothesis.

Introduction

Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the metabolic syndrome and is defined as the accumulation of fat in the liver in patients who do not consume excessive alcohol. The prevalence of NAFLD is 20–30% in adults and is higher in industrialized countries [1]. NAFLD is asymptomatic in most affected patients and is associated with obesity and features of the metabolic syndrome, namely hypertension, dyslipidemia, central adiposity and insulin resistance (IR) or diabetes [2], [3]. The term NAFLD encompasses a wide spectrum of conditions, from simple accumulation of fat (‘fatty liver’ or steatosis) to steatohepatitis (NASH), fibrosis and cirrhosis with its clinical consequences [4].

Despite its high prevalence, only a small proportion of subjects with NAFLD have NASH with consequent higher risk of liver fibrosis, cirrhosis and hepatocellular carcinoma (HCC) (Fig. 1). While patients with simple fatty liver have similar life expectancy to the general population, those with NASH have an impaired survival, due primarily to cardiovascular and liver-related causes. This concept was recently challenged by two studies with longitudinal follow-up, which showed that advanced fibrosis, but not the presence of NASH (as diagnosed by the NAS score) predicted overall mortality in patients with NAFLD [5], [6].

This could be due to shortcomings of the NAS score, as the presence and grading of steatosis have a disproportionate impact compared to lobular inflammation and ballooning, while portal inflammation is not part of the score [7]. Alternatively, lobular inflammation and ballooning could be epiphenomena similarly to simple steatosis, not related to the activation of fibrogenic pathways.

The diagnosis of NAFLD remains one of exclusion and liver biopsy is still the gold standard to differentiate fatty liver from NASH and to stage fibrosis, although several noninvasive markers have been recently introduced for the latter [8].

The underlying mechanism for the development and progression of NAFLD is complex and multifactorial. Different theories have been formulated, leading initially to the ‘two hits hypothesis’. According to this, hepatic accumulation of lipids secondary to sedentary lifestyle, high fat diet, obesity and insulin resistance, acts as the first hit, sensitizing the liver to further insults acting as a ‘second hit’. The ‘second hit’ activates inflammatory cascades and fibrogenesis [9]. This has been supported by animal models of obesity, such as the leptin deficient ob/ob mice, characterized by increased hepatic lipid accumulation, where a second insult is necessary to initiate inflammation and fibrosis [10].

However, it became rapidly evident that this view is too simplistic to recapitulate the complexity of the human NAFLD where multiple parallel factors, acting synergistically in genetically predisposed individuals, are implicated in the development and progression of disease.

Consequently, a multiple-hit hypothesis has now substituted the outdated two-hit hypothesis for the progression of NAFLD. In this review, we explore the potential mechanisms that are implicated in the pathogenesis and progression of NAFLD and that delineate the multiple-hit hypothesis.

Dietary habits, environmental and genetic factors can lead to the development of insulin resistance, obesity with adipocyte proliferation and changes in the intestinal microbiome.

Insulin resistance is one of the key factors in the development of steatosis/NASH and results in increased hepatic de novo lipogenesis (DNL) and impaired inhibition of adipose tissue lipolysis, with consequent increased flux of fatty acids to the liver [11]. Insulin resistance also promotes adipose tissue dysfunction with consequent altered production and secretion of adipokines and inflammatory cytokines [12].

Fat accumulates in the liver in the form of triglycerides, and this happens contemporarily with increased lipotoxicity from high levels of free fatty acids, free cholesterol and other lipid metabolites: as a consequence, mitochondrial dysfunction with oxidative stress and production of reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress associated mechanisms, are activated [13].

Also, altered gut flora leads to further production of fatty acids in the bowel, increased small bowel permeability and thus increased fatty acid absorption and raised circulating levels of molecules which contribute to the activation of inflammatory pathways and release of proinflammatory cytokines such as IL-6 and TNF-α [14].

In subjects predisposed by genetic factors or epigenetic modifications, all these factors affect hepatocyte fat content and liver inflammatory environment, thus leading to a state of chronic hepatic inflammation (Fig. 2) through heterogeneous hepatocellular damage pathways, with possible progression to hepatocellular death (for both direct toxicity and apoptosis activating mechanisms), activation of hepatic stellate cells and deposition of fibrous matrix.

Although the dictum was that steatosis always precedes inflammation, it is now recognized that NASH can be the initial liver lesion: the timing and combination of genetic, external and intracellular events rather than the simple sum of hepatic insults result in different pathways which lead to steatosis or NASH respectively [15].

Section snippets

Fat Metabolism, Lipotoxicity and Insulin Resistance

Fat accumulates in the liver of patients with NAFLD mainly in the form of triglycerides [16], [17]. Triglycerides derive from esterification of glycerol and free fatty acids (FFAs). Once synthesized, triglycerides enter storage or secretory pools, which have distinct rates of turnover. FFAs derive either from diet or from the adipose tissue via lipolysis and/or from hepatic DNL. Once in the hepatocytes, FFAs undergo acyl-CoA synthases activity and form fatty acyl-CoAs, which may enter either

Adipose Tissue Dysfunction

Adipose tissue is not inert as traditionally thought but has an endocrine function and secretes hormones (adipokines) such as leptin and adiponectin [67].

Obesity-related adipocyte hypertrophy and/or insulin resistance result in an imbalance of adipokines that may profoundly affect not only the adipose tissue itself but also the liver [68].

Adipose tissue contributes to the maintenance of low-grade inflammatory states by producing pro-inflammatory cytokines: serum levels of IL-6 and adipocytes

Genetic Determinants

Genetic variants, especially in the form of single nucleotide polymorphisms, influence hepatic FFAs flux, oxidative stress, response to endotoxins and cytokine production and activity, and are determinants of NAFLD development and progression [88].

Genome-wide association studies have established the role of patatin-like phospholipase 3 (PNPLA3) single nucleotide polymorphisms in the development and progression of NAFLD, in particular the I148 M (rs738409 C/G) variant. PNPLA3 gene encodes for a

Epigenetic Factors

Clinical studies investigating epigenetic reprogramming in NASH are just beginning to emerge.

Epigenetic modifications are stable changes at transcriptional level, such as DNA methylation, histones modifications and activity of microRNAs (miRNAs), which do not alter the basic DNA sequences and contribute to the cell homeostasis exhibiting a high degree of developmental and environmentally driven plasticity [100]. It has been hypothesized that the disruption of this balance may determine an

Dietary Factors

Dietary factors, both in terms of quantity and caloric intake but also of specific nutrients, contribute to the development of NAFLD and NASH. In a study of 18 healthy individuals, doubling the regular caloric intake with fast-food based meals resulted in profound ALT elevations and increased steatosis in less than 4 weeks [117].

Fructose is a lipogenic, pro-inflammatory dietary factor that results in oxidative stress and upregulation of TNF-α [118]. It is cleared by the liver within first-pass

Conclusions

The pathogenesis of NAFLD and its progression is a complex process, which cannot be completely explained by the ‘two-hit’ hypotheses. It is becoming increasingly evident that the different histological patterns of NAFLD, namely simple steatosis and NASH not only have different risk of progression but may also reflect different disease entities in terms of pathogenesis. A number of diverse parallel processes contribute to the development of steatosis and liver inflammation. The gut microbiome

Conflicts of Interest

None.

Acknowledgments

Dr. Buzzetti was supported by an educational grant from AIGO (Associazione Italiana Gastroenterologi Ospedalieri) and MIMI (Montecatini Interactive Medicine International).

References (147)

  • A.J. Sanyal et al.

    Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities

    Gastroenterology

    (2001)
  • H. Cortez-Pinto et al.

    Non-alcoholic fatty liver: another feature of the metabolic syndrome?

    Clin Nutr

    (1999)
  • D. Pessayre et al.

    NASH: a mitochondrial disease

    J Hepatol

    (2005)
  • K. Begriche et al.

    Mitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it

    Mitochondrion

    (2006)
  • S. Seki et al.

    Clinicopathological significance of oxidative cellular damage in non-alcoholic fatty liver diseases

    Hepatol Res

    (2005)
  • P. Puri et al.

    Activation and dysregulation of the unfolded protein response in nonalcoholic fatty liver disease

    Gastroenterology

    (2008)
  • A. Kapoor et al.

    Endoplasmic reticulum stress and the unfolded protein response

    Clin Liver Dis

    (2009)
  • J. Crespo et al.

    Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patients

    Hepatology

    (2001)
  • M.F. Gregor et al.

    Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease

    J Lipid Res

    (2007)
  • K. Ikejima et al.

    Leptin augments inflammatory and profibrogenic responses in the murine liver induced by hepatotoxic chemicals

    Hepatology

    (2001)
  • S.S. Choi et al.

    Leptin promotes the myofibroblastic phenotype in hepatic stellate cells by activating the hedgehog pathway

    J Biolumin Chemilumin

    (2010)
  • K. Imajo et al.

    Hyperresponsivity to low-dose endotoxin during progression to nonalcoholic steatohepatitis is regulated by leptin-mediated signaling

    Cell Metab

    (2012)
  • H. Tilg et al.

    Nonalcoholic fatty liver disease: cytokine-adipokine interplay and regulation of insulin resistance

    Gastroenterology

    (2006)
  • Y. Kamada et al.

    Enhanced carbon tetrachloride-induced liver fibrosis in mice lacking adiponectin

    Gastroenterology

    (2003)
  • C. Pirazzi et al.

    Patatin-like phospholipase domain-containing 3 (PNPLA3) I148M (rs738409) affects hepatic VLDL secretion in humans and in vitro

    J Hepatol

    (2012)
  • Y.L. Liu et al.

    Carriage of the PNPLA3 rs738409 C > G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma

    J Hepatol

    (2014)
  • M. Zeybel et al.

    Epigenetic modifications as new targets for liver disease therapies

    J Hepatol

    (2013)
  • P. Angulo

    Nonalcoholic fatty liver disease

    N Engl J Med

    (2002)
  • K. Hassan et al.

    Nonalcoholic fatty liver disease: a comprehensive review of a growing epidemic

    World J Gastroenterol

    (2014)
  • E. Tsochatzis et al.

    Metabolic syndrome is associated with severe fibrosis in chronic viral hepatitis and non-alcoholic steatohepatitis

    Aliment Pharmacol Ther

    (2008)
  • P. Angulo et al.

    Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease

    Gastroenterology

    (2015)
  • M. Ekstedt et al.

    Low clinical relevance of the nonalcoholic fatty liver disease activity score (NAS) in predicting fibrosis progression

    Scand J Gastroenterol

    (2012)
  • E. Buzzetti et al.

    Noninvasive assessment of fibrosis in patients with nonalcoholic fatty liver disease

    Int J Endocrinol

    (2015)
  • W. Peverill et al.

    Evolving concepts in the pathogenesis of NASH: beyond steatosis and inflammation

    Int J Mol Sci

    (2014)
  • E. Bugianesi et al.

    Insulin resistance in nonalcoholic fatty liver disease

    Curr Pharm Des

    (2010)
  • A. Guilherme et al.

    Adipocyte dysfunctions linking obesity to insulin resistance and type 2 diabetes

    Nat Rev Mol Cell Biol

    (2008)
  • Y. Yilmaz

    Review article: is non-alcoholic fatty liver disease a spectrum, or are steatosis and non-alcoholic steatohepatitis distinct conditions?

    Aliment Pharmacol Ther

    (2012)
  • M.M. Jacome-Sosa et al.

    Fatty acid sources and their fluxes as they contribute to plasma triglyceride concentrations and fatty liver in humans

    Curr Opin Lipidol

    (2014)
  • A. Ferramosca et al.

    Modulation of hepatic steatosis by dietary fatty acids

    World J Gastroenterol

    (2014)
  • S.K. Koliwad et al.

    DGAT1-dependent triacylglycerol storage by macrophages protects mice from diet-induced insulin resistance and inflammation

    J Clin Invest

    (2010)
  • K. Yamaguchi et al.

    Inhibiting triglyceride synthesis improves hepatic steatosis but exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis

    Hepatology

    (2007)
  • B.A. Neuschwander-Tetri

    Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites

    Hepatology

    (2010)
  • J. George et al.

    Nonalcoholic fatty liver disease: pathogenesis and potential for nuclear receptors as therapeutic targets

    Mol Pharm

    (2008)
  • J.R. Schultz et al.

    Role of LXRs in control of lipogenesis

    Genes Dev

    (2000)
  • T.C. Schreuder et al.

    Nonalcoholic fatty liver disease: an overview of current insights in pathogenesis, diagnosis and treatment

    World J Gastroenterol

    (2008)
  • N. Stefan et al.

    Causes and metabolic consequences of fatty liver

    Endocr Rev

    (2008)
  • C. Postic et al.

    Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice

    J Clin Invest

    (2008)
  • G.S. Hotamisligil

    Role of endoplasmic reticulum stress and c-Jun NH2-terminal kinase pathways in inflammation and origin of obesity and diabetes

    Diabetes

    (2005)
  • S.E. Shoelson et al.

    Inflammation and insulin resistance

    J Clin Invest

    (2006)
  • G.F. Lewis et al.

    Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes

    Endocr Rev

    (2002)
  • Cited by (1961)

    View all citing articles on Scopus
    View full text