You are on page 1of 17

Gastroenterology 2020;158:1999–2014

MAFLD: A Consensus-Driven Proposed Nomenclature for


Metabolic Associated Fatty Liver Disease

Mohammed Eslam1 Arun J. Sanyal2 Jacob George1, on behalf of the International Consensus Panel

1
Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia;
and 2Virginia Commonwealth University School of Medicine, Richmond, Virginia

Fatty liver associated with metabolic dysfunction is com- reducing disease burden through prevention seems obvious,
mon, affects a quarter of the population, and has no this has not been achieved. Further, although pharmaco-
approved drug therapy. Although pharmacotherapies are therapies are expected to become available in the near
in development, response rates appear modest. The het- future, none to date has been approved. Thus far, several
erogeneous pathogenesis of metabolic fatty liver diseases phase 2b and phase 3 studies either have fallen short of
and inaccuracies in terminology and definitions necessi- meeting current required histologic endpoints, or have done
tate a reappraisal of nomenclature to inform clinical trial so with a modest margin. Muted efficacy of various com-
design and drug development. A group of experts sought to pounds in development are in part a reflection of the
integrate current understanding of patient heterogeneity imprecise definitions and the lack of precision medicine
captured under the acronym nonalcoholic fatty liver dis- including consideration of heterogeneity of the disease.
ease (NAFLD) and provide suggestions on terminology that Despite these alarming data, the nomenclature of the
more accurately reflects pathogenesis and can help in pa-
disease and the criteria for diagnosis have not been updated
tient stratification for management. Experts reached
to reflect our expanding knowledge. The heterogeneity of
consensus that NAFLD does not reflect current knowledge,
the population with NAFLD with respect to its primary
and metabolic (dysfunction) associated fatty liver disease
“MAFLD” was suggested as a more appropriate overarching drivers and coexisting disease modifiers, represent an
term. This opens the door for efforts from the research important impediment to the discovery of highly effective
community to update the nomenclature and subphenotype drug treatments. The phenotypic manifestation of fatty liver
the disease to accelerate the translational path to new diseases likely reflects the sum of the dynamic and complex
treatments. systems-level interactions of these drivers; it follows that
effective treatment requires that they be targeted with
Keywords: MAFLD; Nomenclature; Metabolic; Heterogeneity. precision, based on a person’s phenotype and genetic
background.8,9 However, trial recruitment is currently based
on histologic grading and staging, and that is a problem

S ince the term nonalcoholic fatty liver disease


(NAFLD) was coined by Ludwig and colleagues1 in
1980 to describe fatty liver disease arising in the absence of
because many pathways lead to the same histologic
phenotype, without dissection of the predominant patho-
genic pathways.10,11 Perhaps not surprisingly, the response
significant alcohol intake, the nomenclature and criteria for rates to current investigational agents range from 20% to
a diagnosis has not been revisited. Yet, this disease has risen 40% with a difference from placebo of 10% to 20%.8 Thus, a
in prevalence, with a major impact on clinical and economic “one size fits all approach” would seem inappropriate when
burden to society, such that nearly 1 billion people globally dealing with a very heterogeneous liver disease.
are affected.2 Of concern, NAFLD is increasingly recognized
and diagnosed in children and adolescents,3 and this, when
paired with the intimately associated hepatic as well as Abbreviations used in this paper: BMI, body mass index; lncRNA, long
cardiovascular and oncological sequlae,4,5 places an enor- noncoding RNA; MAFLD, metabolic (dysfunction) associated fatty liver
disease; miRNA, microRNA; NAFLD, nonalcoholic fatty liver disease;
mous burden on individuals, families, and health care sys- NASH, nonalcoholic steatohepatitis.
tems.6 The estimated annual medical costs directly Most current article
attributable to NAFLD exceed V35 billion in 4 large Euro-
© 2020 by the AGA Institute
pean countries (United Kingdom, France, Germany, and 0016-5085/$36.00
Italy) and $100 billion in the United States.7 Although https://doi.org/10.1053/j.gastro.2019.11.312
2000 Eslam et al Gastroenterology Vol. 158, No. 7

From the patient’s perspective, the term “nonalcoholic the suggested terminology. To ensure a robust and transparent
fatty liver disease” not only trivializes the problem by process, anonymity of the participants was maintained.
including terms such as “non,” but is also pejorative, as it
introduces words such as “alcoholic,” potentially placing the
Metabolic Associated Fatty Liver Disease: A
blame on patients as having caused their condition. It also
implies that the treatment must entirely lie in the patient’s
Heterogeneous Phenotype
hand. This has enormous implications on how industry and We now recognize that metabolic fatty liver disease is a
phenotype with complex and disparate causes; the current
policy makers choose to allocate resources for tackling the
terminology (NAFLD) represents an umbrella term for the
syndrome, which clearly is a major cause of death. Lessons
multiple underlying subtypes.17,18 This is evidenced by the
can be learned from cardiologists, diabetologists, neurolo-
wide spectrum of disease severity and natural history, as well
gists, and oncologists who have successfully distanced the as the substantial interpatient variability across the spectrum.
disease they are trying to treat from the underlying obesity, Although hepatic steatosis is highly prevalent, only a minority
smoking, alcohol abuse, and drug abuse. Some of these exhibit inflammatory injury at any time; more importantly, an
factors have high genetic predisposition. In support of this individual can oscillate between steatosis and steatohepatitis
idea, a meeting organized by the European Liver Patient’s even over a short timeframe.19 In addition, although there is
Association with the European Commission in 2018 sug- convincing evidence that liver-related complications (ie,
gested that a change in nomenclature was one of their key cirrhosis and cancer) are more likely in those with steatohe-
requirements. patitis, progression is far from inevitable.19–21 Further, there is
As a first step to tackle this challenge, revising the growing evidence that hepatocellular carcinoma can develop in
nomenclature and definitions of the disease is critical. a fatty liver in the absence of cirrhosis.22 Even among those
Recently, concerns over the inaccuracies of the nomencla- with steatohepatitis, there appear to be individuals with
ture of fatty liver disease have been raised by individual apparent rapid-fibrosis progression and those with inherently
experts.12–14 In prior work, we called for a consensus to slow-fibrosis progression.23 Finally, disease evolution can be
consider these aspects,15 and in this review, an international modified by exogenous interventions (for instance, lifestyle
panel sought to integrate epidemiological knowledge about changes),24 superimposed disease states (eg, type 2 diabetes
disease progression that includes steatosis and steatohe- mellitus),25 inherited predisposition,26 and can even “sponta-
patitis associated with metabolic dysfunction, with infor- neously” regress, as has been demonstrated in placebo group
mation about risk prediction derived from genetic and participants in treatment trials and by observational dual-
phenotyping studies. We suggest a new nomenclature based biopsy studies in secondary/tertiary care settings.23,27,28 Add-
ing to the complexity, it is unknown if the propensity for
on consensus voting by participants to describe the disease
metabolic fatty liver diseases progression can vary across the
that will allow us to properly subphenotype and stratify
life span. For example, given the rapidly escalating prevalence
patients, via the application of more precise genetic,
of metabolic fatty liver disease in children and young adoles-
anthropometric, and metabolic phenotyping approaches. In cents, we still do not understand if their natural history follows
turn, detailed phenotyping will translate into individualized a different trajectory from those who develop disease in
risk prediction and prevention strategies, and improve- adulthood, middle age, or even old age.29
ments in clinical trial design.
Sources of Heterogeneity
The heterogeneity in clinical presentation and disease
Methods course of fatty liver disease is likely influenced by multiple
Following discussions, an initial concept sheet was circu- factors, including age, gender, hormonal status, ethnicity, diet,
lated to the panel of contributors. This revealed widespread alcohol intake, smoking, genetic predisposition, the microbiota,
agreement and consensus that it was time to revisit the and metabolic status. Thus, the final outcome will reflect the
nomenclature of metabolic fatty liver disease as a critical initial balance of these diverse inputs, each interacting with the other
step for improved patient subphenotyping, clinical trials design, and modifying the ultimate manifestations and clinical course
and ultimately, for personalization of medicine. (Figure 1). It follows that effective treatment will require sys-
Subsequently, a manuscript was drafted, circulated to the tematic dissection of the pathways involved and likely multi-
panel, and feedback incorporated over several rounds of revi- faceted and personalized treatments.30,31 A brief summary of
sion. To reach consensus on a nomenclature, the Delphi method current knowledge about factors contributing to NAFLD het-
was adopted in 2 rounds. This method is a recommended erogeneity is provided below.
iterative process for use in the health care setting as a reliable
means to solicit and distil the judgments of experts and to
determine consensus via a systematic progression of repeated Age and Gender
rounds of voting.16 A “closed” electronic survey URL was sent NAFLD prevalence, the risk of hepatic and extrahepatic
to participants providing a unique link that could be used only complications, and the likelihood of overall and disease-specific
once. Survey data were collected and managed using REDCap mortality increases with advancing age.19,21,32,33 With aging,
(Research Electronic Data Capture). In the first round of sur- substantial changes occur in the liver, including a decline in
veys members suggested 1 or more terms to describe metabolic hepatic blood flow, hepatic volume, and liver function, a
fatty liver disease. In a second round (based on a summary of reduction in bile acid synthesis and alterations in cholesterol
the experts’ suggestions), participants were asked to vote on metabolism, as well as a reduction in mitochondrial number
May 2020 MAFLD: A Proposed Nomenclature 2001

with subsequent increases in oxidative respiration.34 Cellular demonstrated sexual differences in liver gene expression of
senescence has also been implicated.35,36 Furthermore, aging is regulators of multiple metabolic pathways using a mice
accompanied by changes in body composition, including a computational model. Notably, some such as peroxisome
decrease in muscle mass, an increase in abdominal adiposity proliferator-activated receptor PPARa, farnesoid X receptor,
and ectopic fat deposition, with increases in insulin resistance and liver X receptor, which are highly gender dependent, are
and prevalence of the metabolic syndrome.37,38 Emerging evi- currently being investigated as therapeutic targets for stea-
dence suggests that sarcopenia is associated with both NAFLD tohepatitis.51 A further study demonstrated gender-related
and NAFLD-related advanced fibrosis, even after adjusting for pathways contribute to steatosis and fibrosis in male and fe-
body mass index (BMI) and insulin resistance.39,40 Presumably, male mice (males mainly inflammation and females mainly
aging also captures greater exposure to the drivers, which alterations of redox state), despite similar endpoints.52
result in steatohepatitis and fibrosis. Clearly, sex and menopausal status influence disease out-
Equally, as recently reviewed,41 there is substantial sex- comes and require stratification as treatment responses can
ual dimorphism in many aspects of fatty liver disease with vary substantially.
regard to risk factors, prevalence, fibrosis pattern, and dis-
ease outcomes. Generally, prevalence tends to be lower in
women predominantly at earlier disease stages, whereas Ethnicity
disease frequency increases in postmenopausal women.41 Population-based data show ethnic differences in the
Similarly, fatty liver prevalence is lower in post-menarchal prevalence of fatty liver; a recent meta-analysis demonstrated
girls than in boys.42 Among postmenopausal women, those both NAFLD prevalence and risk of nonalcoholic steatohepa-
not on hormone replacement therapy tend to have higher titis (NASH) were highest in Hispanic individuals, intermedi-
disease prevalence compared with those on hormone ate in white individuals, and lowest in black individuals;
replacement therapy,43 and similarly, premenopausal women however, fibrosis risk did not differ according to ethnicity.53
have less severe hepatic fibrosis and better survival Metabolic fatty liver disease is also rapidly increasing in
compared with men and postmenopausal women.44,45 Asian populations.54 Previous studies have demonstrated that
Consistently, a longer duration of estrogen deficiency asso- Asian individuals tend to accumulate liver fat at lower BMI
ciates with a higher likelihood of fibrosis among post- compared with those of other races.55 The course of disease
menopausal women with fatty liver disease.46 By analogy, also appears to be more severe in Asian compared with non-
studies of diet-induced mouse models suggests that males Asian individuals, and they tend to have more lobular
develop more severe steatosis and liver histology compared inflammation and higher grades of ballooning compared with
with females.47,48 other ethnicities.56,57 Although data regarding fibrosis are
Although the mechanisms for these effects are not scarce in the preceding studies, Asian individuals tended to
completely understood, sex differences in adiposity, metabolic have a higher risk of fibrosis, whereas African individuals
risk factors, and body fat distribution (which tends to shift were at lower risk compared with white individuals; this did
toward abdominal obesity after menopause), likely play a not reach significance, perhaps due to sample size limita-
role.49 A recent study in mice from approximately 100 strains tions.56,57 However, and notably, these biopsy-based studies
included in the hybrid mouse diversity panel demonstrated might suffer from selection bias. For example, a community-
that multiple molecular pathways and gene networks impli- based study in Hong Kong suggested that although NAFLD is
cated in lipid metabolism, insulin-signaling, and inflammation detected in a quarter of the population, the prevalence of
show sexual dimorphism.50 Similarly, another study advanced fibrosis is low.58

Figure 1. Heterogeneity of
MAFLD. The heterogeneity
in clinical presentation and
course of fatty liver dis-
ease is influenced by a
multitude of factors,
including age, sex,
ethnicity, alcohol intake,
dietary habits, hormonal
status, genetic predispo-
sition, and epigenetic fac-
tors, the microbiota, and
metabolic status. It is likely
that there is a differential
impact in the contribution
of the various factors in
any individual over time
and among individuals that
then shapes disease
phenotype and course.
2002 Eslam et al Gastroenterology Vol. 158, No. 7

The reasons for racial disparities in fatty liver risk are not Light and Moderate Alcohol Use
completely understood. Plausible explanations include varia- Since its first description, metabolic has been considered
tions in genetic predisposition, metabolic attributes, cultural distinct from alcoholic-associated liver disease based on a
and socioeconomic factors, dietary and exercise habits, and cutoff of daily alcohol intake of 30 g daily for men and 20 g
access to health care, as well as environmental risks. There are daily for women. The assumption underlying the cutoff has
substantial differences in genetic heritage across ethnic been that alcohol intake below these thresholds does not
groups; variation in the risk allele of the Patatin-like phos- induce hepatic steatosis or have deleterious impacts on liver
pholipase domain-containing protein 3 (PNPLA3) gene that is disease progression and outcomes.65
most frequent in Hispanic individuals (49%), followed by non- Because of the high prevalence of adults with NAFLD who
Hispanic white (23%) and African American individuals drink at least in moderation (w4 drinks/week),66 there is now
(17%) has helped, at least partially, to explain some of this much interest in the influence of light and moderate alcohol use
ethnic variability.59–61 In addition, the risk allele of the on the prognosis of NAFLD, with debate on the protective ef-
PNPLA3 rs738409 polymorphism was found to be more fects67,68 and perceived harms.69,70 More recently, there has
common in East Asian than Caucasian individuals.62 Notably, been evidence for and against safe limits for alcohol con-
because the effect size of fatty liver–related gene variants sumption in the setting of NAFLD.71 Some reports suggest that
supports the existence of differences among races, the relative modest alcohol consumption, even after adjustment for previ-
contribution of specific genetic and and environmental trig- ous heavy drinking, is associated with a reduction in vascular
gers (eg, dietary factors) or modifying risk variants, toward complications67,68 or has no impact.72 Other studies have
disease pathogenesis is likely variable among ethnic groups demonstrated that moderate drinking (2 drinks a day for
(Figure 2). women and 3 drinks a day for men) is associated with a
On the other hand, there are marked racial/ethnic so- reduced prevalence of NASH and advanced fibrosis.73 In
cioeconomic disparities that are likely also reflected in dif- contrast, some studies highlight that even low alcohol intake in
ferences in multiple disease risk factors. For example, there those with a fatty liver is associated not only with increased
is a clear difference between European and Asian pop- risk of disease progression, but also for advanced liver disease
ulations with regard to insulin resistance and body fat dis- and cancer,74–77 and decreased rates of improvement in stea-
tribution, as discussed later. There are also disparities in tosis and NASH.78 The effect of alcohol use on liver disease
physical activity; in 2016, a report including 1.9 million evolution likely has a dose-response, rather than a J-shaped
participants across 168 countries suggested that women in association,79,80 with a synergistic detrimental effect with the
Latin America, south Asia, and high-income Western coun- presence of metabolic syndrome,74,76 as has recently been
tries have the highest prevalence of physical inactivity.63 reviewed.73
Likewise, data from the NASH Clinical Research Network
reported less physical activity, increased carbohydrate con-
sumption, and lower income levels in Hispanic individuals Dietary Intake, Gut Microbiota, and Bile Acids
compared with non-Hispanic white patients with NASH.64 A For metabolic homeostasis, the neuroendocrine axis, di-
role for gut microbiota could also be implicated, as discussed etary intake, muscle mass, physical activity, and the enter-
later in this article. ohepatic circulation, gut microbiota, bile acids, and their

Figure 2. Interindividual variation in the predominant drivers of MAFLD. MAFLD is a complex phenotype shaped by the dy-
namic interaction of genetic predisposition with environmental factors and components of the metabolic syndrome. The effect
size of genetic variants and the predominant drivers can exhibit marked interindividual variation. As an example, disease in
patient 1 is driven predominantly by environmental influences with less contribution from genetic predisposition; in patient 2,
metabolic syndrome is the predominant driver, whereas disease in patient 3 is driven by genetic factors with a limited
contribution from other factors. Identification of the predominant drivers in every patient can help in personalization of
medicine.
May 2020 MAFLD: A Proposed Nomenclature 2003

related metabolites are intimately implicated in fatty liver Current consensus suggests that the distribution and the
pathogenesis (Supplementary Figure 1). The dietary pattern overall health of fat, rather than its amount is likely the major
that characterizes the Western diet, including increased fat determinant of disease risk. For example, higher amounts of
and fructose consumption that is fueling the increase in visceral relative to peripheral and subcutaneous adipose tissue
obesity and fatty liver, is associated with a wide range of is associated with greater metabolic risk108,109 and is directly
metabolic dysfunction, including insulin resistance and linked to liver inflammation and fibrosis, independent of insulin
abnormal lipid profile.81 In contrast, adoption of a Mediter- resistance and hepatic steatosis.108 Sex, sex hormones (as dis-
ranean dietary pattern is accompanied with a decrease in liver cussed previously), ethnicity, and genes obviously play impor-
fat in patients with NAFLD and a decrease in cardiovascular tant roles in determining the location and health of adipose
risk.82,83 tissue. There is, for example, strong evidence that ethnicity is
Microbiota composition can change rapidly and widely ac- implicated in determining fat distribution and health.110 Thus,
cording to dietary patterns82,84 and the involvement of the gut abdominal and visceral adiposity are greater among Asian in-
microbiome in fatty liver and steatohepatitis in both mice and dividuals compared with Caucasian individuals, and lower in
humans is well recognized.85,86 Emerging data suggest that the African individuals,111–114 as is insulin resistance despite an
microbiome and gut microbiome–derived metabolites can equal or lower BMI.115–117 Genetic variants also play a role in
predict advanced fibrosis and cirrhosis in NAFLD.87–90 Gut the regulation of fat distribution,118–120 with “favorable
microbiota are also implicated in regulating bile acids and their adiposity” genes having been recently identified.102,121,122
metabolites, which in turn regulate glucose, lipid, and choline Although lipid accumulation in liver is a hallmark of NAFLD,
metabolism, and energy homeostasis.91 Altered gut flora and there is emerging evidence that there is likely a variety of un-
intestinal permeability have also been shown in patients and derlying mechanisms and routes for its development. For
murine models of NAFLD.92,93 This leads to increased circu- instance, a recent study has demonstrated that lipid composi-
lating levels of bacterial products including lipopolysaccharide, tion in liver is very different in 2 proposed subtypes of NAFLD.
as well as other bioactive compounds that may induce intra- In subtype 1, based on insulin resistance, patients tend to have
hepatic activation of proinflammatory cells, hepatic stellate monounsaturated triacylglycerols and free fatty acids enriched
cells, and hepatocytes via stimulation of toll-like receptors with ceramides in liver, whereas subtype 2, based on carrying
(particularly 2, 4, and 9), a sensor for these products.94–96 the PNPLA3 risk genotype at rs738409, have polyunsaturated
However, it remains challenging to disentangle the effects of triacylglycerols.123 Similarly, another study suggested the ex-
diet and its associated consequences for liver disease, from istence of 3 NAFLD subtypes, with different metabolic pheno-
effects mediated by diet-induced alterations to the microbiome, types.124 In another study, regions with steatosis demonstrated
and to ascertain causality under these same conditions. distinct lipid composition, predominantly in the form of a loss
Notably, a role for human genetic variation and ethnicity in of arachidonic acid-containing intracellular phospholipids,
driving differences in microbiomes has recently been sug- compared with nonsteatosis liver tissue.125 A further report
gested.97–100 used RNA-sequencing analysis and identified molecular sub-
types with distinct gene expression pattern clusters that are
implicated in lipid metabolism, interferon signaling, and im-
Obesity and Metabolic Health mune system pathways, according to different histological
Although obesity intimately associates with liver fat, not all scores.126 In total, these new datasets emphasize that there are
patients with obesity develop metabolic fatty liver disease.2 likely multiple NAFLD subtypes characterized by unique
Whereas obesity can be classified as metabolically healthy metabolomic signatures. Based on subtype, it is likely that
obesity and metabolically unhealthy obesity, with the former treatment responses will vary, and hence defining the meta-
affecting approximately 45% of obese subjects, there is no bolic landscape of an individual is likely important in clinical
consensus on a definition of metabolic health. Various defini- trial design.
tions of metabolic syndrome include a combination of different
metabolic components.101,102 Similarly, although insulin resis-
tance is believed to play a pivotal role and is a pathophysio- Lean NAFLD
logical feature of fatty liver,103 it has not been included in Currently, lean NAFLD, or NAFLD in lean individuals, is
several definitions of metabolic syndrome. Notably, multiple defined as hepatic steatosis with a BMI <25 kg/m2 (or
large-scale cohort studies do not clearly support the notion that <23 kg/m2 in Asian individuals) in the absence of “significant”
metabolically healthy obesity subgroups, at least as currently alcohol intake.127 Although first described in Asian pop-
defined, are protected from cardiometabolic complications ulations, it is recognized that between 5% and 45% of patients
compared with those with a stable normal weight who are with NAFLD are lean; even among European individuals,
metabolically healthy.104–106 Better classification based on approximately 20% of patients are considered lean.128
molecular or genetic profiling could help dissect with high Although those with lean NAFLD have a better metabolic
precision, metabolically favorable and unfavorable subtypes, and histological profile compared with their counterpart
with distinct metabolism, anthropometry, and patterns of fat obese subjects, their natural history is poorly defined, with
deposition, and likely differential responses to drug treat- some data suggesting they may have a worse outcome and
ments.107 On the other hand, approximately 30% of normal- accelerated disease progression,129,130 whereas others sug-
weight individuals can be classified as metabolically obese gest no difference or even better outcomes.131,132 More recent
normal weight, and they demonstrate an increased propensity data propose that lean NAFLD comprises a distinct patho-
for cardiometabolic risk; a fair proportion of patients with a physiological entity from that in obese subjects, which extends
fatty liver are also lean. beyond just simple differences in BMI. In that study, lean
2004 Eslam et al Gastroenterology Vol. 158, No. 7

patients had distinct metabolic and gut microbiota profiles and advanced fibrosis,151–153 even in those with viral hepa-
compared with their obese counterparts and lean healthy titis.154 Although early reports suggested that PNPLA3
controls. Specifically, they had intact metabolic adaptation in rs738409 has no association with the metabolic profile,155
response to an obesogenic environment via increased bile more recent larger studies and a Phenome-wide association
acids and farnesoid X receptor activity that likely helped them study (PheWAS) study indicate that it has similar metabolic
to maintain an obesity-resistant phenotype. Notably, either effects to TM6SF2 rs58542926.156,157 An association of
this adaptation tends to be lost with advancement of disease PNPLA3 rs738409 and TM6SF2 rs58542926 with type 2
or the failure to adapt promotes disease progression. Other diabetes has also been demonstrated beside the known as-
intriguing aspects from a subset of the patients suggests that sociation of GCKR rs1260326 with diabetes.158 Variants in
they have a distinct gut microbiota profile, with enrichment of HSD17B13 and MBOAT7 do not to date appear to have an
species implicated in the generation of liver fat, and a genetic effect on serum lipids, glycemia, or risk of coronary heart
profile with an increased prevalence of the TM6SF2 risk disease.159–162
allele,133 as also observed by another study.134 Further
studies will be required to explore whether the metabolic Epigenetic Factors
adaptation observed in lean NAFLD is seen in other subtypes Reversible epigenetic changes represent a plausible bridge
of patients. between genes and the environment; their dysregulation is
implicated in several diseases, including NAFLD.139 Numerous
Familial Risk microRNAs (miRNAs) have been linked to NAFLD. A recent
meta-analysis demonstrated that in particular, miRNA-122,
Data from well-characterized cohorts of twins who un-
miRNA-34a, and miRNA-192 could be biomarkers of fatty
derwent imaging to quantify liver fat and fibrosis have shown
liver disease.163,164 miRNA-122 and miRNA-192 showed upre-
that both are heritable traits.135 Furthermore, retrospective
gulation in NAFLD compared with healthy controls, whereas
family-based studies show that there is familial aggregation of
miRNA-34a was upregulated in NAFLD and correlated with
NAFLD and cirrhosis.136 Consistently, a recent prospective
disease severity.163,164
study including probands with NAFLD-cirrhosis and their
Data on the role of long noncoding RNAs (lncRNAs) and
first-degree relatives indicated that the risk of advanced
other type of noncoding RNAs in NAFLD is limited. Some data
fibrosis among first-degree relatives of patients with cirrhosis
suggest alterations in lncRNAs in NASH, such as a hepatic-
is 18%.137 This is substantially higher than the risk of
specific lnc18q22.2,165 a brown fat-enriched lncRNA 1
cirrhosis in the general population and points toward further
(Blnc1),166 and metastasis-associated lung adenocarcinoma
substratification of the population by family history of
transcript 1.167 A study using genome scanning with next
cirrhosis due to NAFLD.
generation sequencing has identified other candidates.168 The
role of lncRNAs in steatohepatitis remains to be further eluci-
Genetic Variation dated in larger cohorts.
Genome-wide association and large candidate studies have Several studies show wide alterations in the methylation
identified multiple loci associated with NAFLD and NASH. signature of hepatic as well as peripheral blood-derived DNA,
Although in-depth discussion is beyond our scope, the topic including regulatory loci for key metabolic, inflammatory,
has recently been reviewed.138,139 At least 5 common variants and fibrotic pathways, in patients with NAFLD. Some of
in different genes have been associated with NAFLD, namely these signatures appear to reverse following bariatric sur-
PNPLA3, transmembrane 6 superfamily member 2 (TM6SF2), gery.169–171 There is also evidence that DNA methylation can
glucokinase regulator (GCKR), MBOAT7, and hydroxysteroid be a biomarker for fibrosis stratification in NAFLD172 and
17-beta dehydrogenase-13 (HSD17B13).139 Multiple other that it regulates the expression of PNPLA3.173 For example,
genes have reported associations, including polymorphisms in hypermethylation of the PPARg promoter can be used to
inflammatory, immune and metabolism-related, oxidative identify patients with advanced fibrosis.172 More recently, a
stress, adipokine, and myokine-related genes.138–143 It is series of studies have shown evidence of methylation of the
noteworthy, however, that all known variants explain only a key mitochondrial urea cycle enzymes carbamoyl phosphate
small proportion of NAFLD, suggesting the existence of heri- synthase-1 and ornithine transcarbamylase enzymes result-
tability factors that are yet to be defined.144 Exploring the role ing in a reduction in their function and hyperammonemia in
of other types of genetic variation, gene-gene and gene- patients with NAFLD.174 Hyperammonemia activates stellate
environment interactions, epigenetics, common variants that cells and is associated with progression of fibrosis in
do not reach genome-wide significance, and rare and less NAFLD175,176; treatment of hyperammonemia using ornithine
common variants will help dissect the missing heritabili- phenylacetate prevented progression of fibrosis in an animal
ty.139,145,146 For example, a gene-environment interaction has model, suggesting a potential novel metabolic therapeutic
been proposed for the PNPLA3 variant with dietary pat- strategy.177
terns,147 increased intake of sugars,148 omega-6 poly- Importantly, epigenetic mechanisms play a crucial role in
unsaturated fatty acids intake,149 obesity, and insulin fetal metabolic programming of liver fat,178,179 with growing
resistance.150 evidence that the earliest origins of NAFLD extend to in
Of interest, described NAFLD-related variants show utero experiences. Data from animals suggest that a
divergent metabolic effects. Multiple reports indicate an as- maternal diet high in fat triggers widespread epigenetic al-
sociation of a genetic variant of TM6SF2 (encoding terations in fetal hepatic DNA, accompanied by metabolic
p.Glu167Lys) with lower serum lipid levels and lower risk of maladaptation that favors an increase in the risk of developing
coronary artery disease, but with increased risk of fatty liver NAFLD in the offspring.180,181 Even paternal diet patterns and
May 2020 MAFLD: A Proposed Nomenclature 2005

prediabetes increase the risk of diabetes in offspring.182 (Mat1a)-deficient mouse can recapitulate a subtype of
Notably, these changes can be transmitted over generations, human NAFLD,124 whereas mice fed a high-cholesterol or
but can also be altered by exercise and lifestyle inter- methionine/choline-deficient diet seem to recapitulate
ventions.183–185 Although data in humans are still limited, several features of lean NAFLD.193 Despite the range of
maternal obesity and patterns of infant nutrition are risk available models, there remains a need to develop
factors for the development of NAFLD in adolescence and improved in vitro and in vivo model systems.
adulthood. For instance, normal pregestational BMI and breast-
feeding for more than 6 months reduces the risk of developing
NAFLD in the mother during mid-life186 and during adoles- Impact on Clinical Trials Design and the Ability to
cence in offspring.187 Similarly, an increase in methylation of Find Treatments
the peroxisome proliferator-activated receptor g coactivator 1 The growing magnitude of NAFLD and the lack of
(PGC1) gene that controls several aspects of energy meta- effective drug treatments is reflected in intense clinical trial
bolism in liver188 and in newborns, is correlated with activity that has jumped from just 8 in 2013 to more than
increased maternal pregestational BMI.189 300 ongoing in 2018.194 Unfortunately, response rates
remain modest, with <20% to 30% of participants
demonstrating NASH resolution and fibrosis regression.
Why Do We Need to Consider NAFLD This low response can be attributed to many factors,
Heterogeneity in Clinical Practice? including heterogeneity in population selection, lack of
Impact on the Performance of Noninvasive stratification based on the underlying dominant driver
mechanisms, and the Hawthorne (placebo) effect.8 There-
Assessment of Fibrosis
fore, the standard clinical trial design that does not take into
Noninvasive fibrosis scores are commonly used to
consideration disease heterogeneity may not be the best
identify or exclude significant or advanced fibrosis in pa-
option for studying a complex disease. Thus, future clinical
tients with fatty liver disease. However, a recent study
trials will likely target patients with specific characteristics
suggested that the performance of scores such as the
(sex, hormonal status, genetic predisposition, metabolic and
NAFLD fibrosis score and fibrosis 4 may vary across the
microbiota signatures, and the presence or absence of co-
life span, with lower specificity among older adults and
morbid conditions) once the relationships between the
lower accuracy in young adults.190 The performance of
characteristics and the treatment targets are understood.
noninvasive scores and the used Transient Elastography
Such trial design will likely include rational combination
liver stiffness cutoffs in different ethnic populations and in
approaches.31
special subpopulations such as diabetic and obese in-
Considering alternative innovative trial designs might be
dividuals also need to be considered. For example, it has
a viable option (Figure 3). Recently, using overarching or
been shown that blood biomarkers are less accurate in
master protocols designed to address multiple questions by
South Asian compared with European individuals, regard-
investigating different drugs (more than 1 or 2 therapies
less of metabolic indices.191 As it is likely that blood-based
that might even include direct comparisons of competing
biomarkers or imaging techniques will supplant liver bi-
drugs) in different conditions (more than 1 patient type or
opsy for the diagnosis of disease in patients who would
disease), all within the same overall trial structure has been
benefit from drug treatment, equally it implies that any
suggested.195 Adaptive trial designs that provide flexibility
future marker should be validated in more precisely
for altering 1 or more aspects of the basic features of the
defined cohorts. Thus, the consensus group suggests that
study design based on responses in earlier phases is also an
the factors that shape the heterogeneity of NAFLD be
option,196 although this will add substantial complexity to
considered when devising and applying risk-stratification
data interpretation. Notably, given the heterogeneity of
scores and algorithms. This approach will continue to
NAFLD according to ethnicity and geographic region,
evolve as new contributors to disease variability are
regional stratification or performing separate trials in
identified.
different geographic regions should be considered for key
trials.
Impact on the Development of Clinically Relevant
Animal Models
The complexity of human NASH is paralleled by the Is NAFLD the Right Name for Metabolic
heterogeneity of animal models and the inability of these Liver Disease?
models to replicate the gamut of disease.192 This repre- How do the preceding considerations influence our
sents both a barrier to the development of novel thera- thinking on the need to revise the definition and nomen-
peutics but also an opportunity to better understand clature for NAFLD? It is clearly the time to do this. The
steatohepatitis pathogenesis based on different drivers of suggestion of this consensus focuses on 4 aspects.
disease. Considering that NAFLD as described today is First, NAFLD was described as a condition of “exclu-
not a single entity, exploring the overlapping features of sion,” which means that it exists only when other con-
preclinical models with subtypes of NAFLD may help in ditions, such as viral hepatitis B and C, autoimmune
overcoming these challenges. For instance, it has been diseases, or alcohol intake above a particular threshold,
reported that the methionine adenosyltransferase 1A are absent. However, with advancements in our
2006 Eslam et al Gastroenterology Vol. 158, No. 7

Figure 3. Innovative clinical trials for MAFLD. The substantial heterogeneity of patients with MAFLD and the limited responses
to investigational targets in current clinical trials imply that innovative trial designs are required. Trial designs such as umbrella,
basket, and adaptive designs have been suggested to overcome the challenges; however, such designs add complexity to the
trial analysis.

understanding of the underlying pathological processes, Third, although in clinical practice we segregate pa-
it is clearly a disease that must be defined by inclusion, tients into those with NASH and those without, whether
rather than by exclusion. Further, given its high preva- this is appropriate is a matter of debate. As we know, there
lence in most affluent populations, especially those is tremendous plasticity in metabolic liver disease over the
consuming a westernized diet, fatty liver disease is life span and strong evidence that fibrosis is the major
recognized to coexist with other conditions such as viral determinant of adverse outcomes.21 Hence, the current
hepatitis, autoimmune diseases, and alcohol,197–199 and classification may be misleading and perhaps metabolic
will have synergistic effects on disease progression.200,201 dysfunction associated fatty liver disease should be
The nomenclature for fatty liver disease and criteria for considered similar to other chronic liver diseases with
diagnosis need to reflect this new knowledge. some degree of activity and a stage of fibrosis, without
Second, there remains debate about the safe limit of dichotomous stratification into NASH and non-NASH. From
alcohol intake. Updating a diagnosis of NAFLD to zero or a pathological perspective, this will result in improved
near to zero alcohol consumption as has been suggested disease classification, at least in the context of liver
by some is clearly impractical, as recently discussed.15 biopsy.26
Furthermore, there are significant methodological chal- Fourth, the heterogeneous nature of fatty liver diseases
lenges in questionnaires used for measuring alcohol suggests that they cannot be considered or managed as a
consumption including documenting prior and over-life single condition with a “one size fits all” approach to ther-
use, low amounts of intake, patient underreporting, and apy. Lack of consideration of heterogeneity impacts and
recall bias, as well as marked variability in defining terms detracts from our ability to precisely define the natural
such as “social drinking” and “binging” in individuals with history of fatty liver phenotypes, to appropriately select for
NAFLD. Thus, linking metabolic fatty liver disease, a clinical trials that are weighted to demonstrate meaningful
distinct entity, to alcohol in its name is problematic. benefits, and to compare or pool results from the trials. For
Moreover, including the term “nonalcoholic” in the name these reasons, an updated and appropriate nomenclature
is disappointing for abstemious patients and links this for the disease is the first step in the long path to decon-
entity to the stigma of alcohol consumption. Confounding volution of disease heterogeneity.
terms in the name of these diseases should be replaced as Based on the preceding, participants agreed on the
has already been done with primary biliary cirrhosis need for a revised and updated terminology; the bulk of
becoming primary biliary cholangitis, with sometimes respondents in the first round of survey suggested that
redundant but more accurate and clear words, defining the words metabolism, fat, and liver be included in some
the entity.202 More importantly, there is an urgent need to form in the name. The final vote favored Metabolic
identify coexisting metabolic and alcohol liver disease so Associated Fatty Liver ± Disease (MAFL/MAFLD) (sup-
that they may be treated appropriately. This group of ported by 72.4% of participants). The second preference,
patients is distinct from those with pure or predominant Metabolic Fatty Liver ± Disease (MEFL/MEFLD), was
alcoholic cirrhosis. Such patients are currently excluded supported by 17.2% (Supplementary Table 1). Thus, the
from all NASH trials. panel suggests we eliminate the term “NAFLD” from the
May 2020 MAFLD: A Proposed Nomenclature 2007

Table 1.Statements of the Consensus Panel


Nomenclature and definition of metabolic associated fatty liver
disease (MAFLD)
 We suggest that the nomenclature of NAFLD should be updated to
MAFLD.
 The diagnosis of MAFLD should be based on the presence of
metabolic dysfunction not the absence of other conditions.
 MAFLD can coexist with other liver diseases.
 A reference to alcohol should not be included in the MAFLD
acronym.
 Patients with both MAFLD and a contribution from alcohol to their
liver disease represent a large and important group that requires
further investigation and characterization.
MAFLD heterogeneity
 MAFLD is a heterogeneous entity.
 Appropriate patient stratification must be considered when
noninvasive fibrosis scores are developed and in clinical trial
design.
 Studies are required to map the landscape of MAFLD and to pre-
cisely define subtypes of the disease.
Clinical trials for MAFLD
 Detailed patient stratification and tailoring clinical trial inclusion
criteria based on drivers of disease will likely yield more informative
and meaningful results.
 Innovative designs for clinical trials and personalized combination
therapy approaches will likely be required to overcome the chal-
lenges of disease heterogeneity and for optimal clinical efficacy. Figure 4. Implications of the proposed update to the MAFLD
nomenclature. The growing burden of MAFLD in the absence
of effective therapies requires an updated process map to
address the challenges. The first step is an update of
lexicon and replace it with metabolic associated fatty liver nomenclature, as without precise terminology, neither patient
“MAFLD.” The term MAFLD represents the overarching care nor science can be adequately served. This update of
umbrella of the common disease we treat and will have nomenclature we expect will be a step toward further char-
multiple subphenotypes, reflecting the dominant driver of acterization of disease heterogeneity. In turn, detailed phe-
notyping can guide the development of better preclinical
disease. Obviously, many, if not most, patients will have models and identify novel therapies that are likely to be
overlapping contributions from other and distinct liver effective for particular patient subtypes, but not others. This
diseases that range from alcohol (regardless the amount) will lead to improved clinical trial designs, allowing us to
to viral hepatitis. The natural history of these latter compare and pool results and thereby help reduce the impact
groups is likely very different from those with pure of disease burden.
metabolic dysfunction.

design of more appropriate clinical trials and for patient


Conclusion management and to consider the implications of the upda-
The outdated NAFLD/NASH acronyms, the criteria for ted nomenclature on clinical practice and public health
diagnosis, and a lack of adequate consideration of hetero- policy (Figure 4).
geneity in risk profiles and treatment responsiveness
represent barriers that hamper progress toward effective
treatments. The consensus group has suggested an acronym Supplementary Material
(MAFLD) that we believe more accurately reflects current Note: To access the supplementary material accompanying
knowledge of fatty liver diseases associated with metabolic this article, visit the online version of Gastroenterology at
dysfunction that should replace NAFLD/NASH. In addition, www.gastrojournal.org, and at https://doi.org/10.1053/
we have identified gaps in current knowledge and highlight j.gastro.2019.11.312.
new strategies and tools to overcome the challenges
(Supplementary Table 2). A summary of suggestions is
provided in Table 1. The group acknowledges the many References
investigators in the field who have made similar well- 1. Ludwig J, Viggiano TR, McGill DB, et al. Nonalcoholic
reasoned pleas for a change in nomenclature. This work steatohepatitis: Mayo Clinic experiences with a hitherto
also opens up for wider consultation with the public, pa- unnamed disease. Mayo Clin Proc 1980;55:434–438.
tients, regulators, and nonhepatology health care workers, 2. Younossi Z, Anstee QM, Marietti M, et al. Global burden
the necessity for a nomenclature update. Future studies will of NAFLD and NASH: trends, predictions, risk factors
allow us to further characterize and subphenotype the dis- and prevention. Nat Rev Gastroenterol Hepatol 2018;
ease and its drivers as a necessary prerequisite for the 15:11–20.
2008 Eslam et al Gastroenterology Vol. 158, No. 7

3. Wiegand S, Keller KM, Robl M, et al. Obese boys at 21. Vilar-Gomez E, Calzadilla-Bertot L, Wong VWS, et al.
increased risk for nonalcoholic liver disease: evaluation Fibrosis severity as a determinant of cause-specific
of 16 390 overweight or obese children and adolescents. mortality in patients with advanced nonalcoholic fatty
Int J Obes (Lond) 2010;34:1468–1474. liver disease: a multi-national cohort study. Gastroen-
4. Jepsen P, Vilstrup H, Mellemkjaer L, et al. Prognosis of terology 2018;155:443–457.e17.
patients with a diagnosis of fatty liver - A registry-based 22. Mittal S, El-Serag HB, Sada YH, et al. Hepatocellular
cohort study. Hepatogastroenterology 2003;50:2101– carcinoma in the absence of cirrhosis in United States
2104. veterans is associated with nonalcoholic fatty liver dis-
5. Sorensen HT, Mellemkjaer L, Jepsen P, et al. Risk of ease. Clin Gastroenterol Hepatol 2016;14:124–131.e1.
cancer in patients hospitalized with fatty liver - A Danish 23. Singh S, Allen AM, Wang Z, et al. Fibrosis progression in
cohort study. J Clin Gastroenterol 2003;36:356–359. nonalcoholic fatty liver versus nonalcoholic steatohepa-
6. Younossi Z, Tacke F, Arrese M, et al. Global perspec- titis: a systematic review and meta-analysis of paired-
tives on nonalcoholic fatty liver disease and nonalcoholic biopsy studies. Gastroenterology 2014;146:S947–S948.
steatohepatitis. Hepatology 2019;69:2672–2682. 24. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L,
7. Younossi ZM, Blissett D, Blissett R, et al. The economic et al. Weight loss through lifestyle modification signifi-
and clinical burden of nonalcoholic fatty liver disease in cantly reduces features of nonalcoholic steatohepatitis.
the United States and Europe. Hepatology 2016; Gastroenterology 2015;149:367–378.e5.
64:1577–1586. 25. Loomba R, Abraham M, Unalp A, et al. Association be-
8. Friedman SL, Neuschwander-Tetri BA, Rinella M, et al. tween diabetes, family history of diabetes, and risk of
Mechanisms of NAFLD development and therapeutic nonalcoholic steatohepatitis and fibrosis. Hepatology
strategies. Nat Med 2018;24:908–922. 2012;56:943–951.
9. Eslam M, George J. Genetic insights for drug develop- 26. Pelusi S, Cespiati A, Rametta R, et al. Prevalence and
ment in NAFLD. Trends Pharmacol Sci 2019;40:506– risk factors of significant fibrosis in patients with nonal-
516. coholic fatty liver without steatohepatitis. Clin Gastro-
10. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit enterol Hepatol 2019;17:2310–2319.e6.
pathogenesis of non-alcoholic fatty liver disease 27. Han MAT, Altayar O, Hamdeh S, et al. Rates of and
(NAFLD). Metabolism 2016;65:1038–1048. factors associated with placebo response in trials of
11. Skoien R, Richardson MM, Jonsson JR, et al. Hetero- pharmacotherapies for nonalcoholic steatohepatitis:
geneity of fibrosis patterns in non-alcoholic fatty liver systematic review and meta-analysis. Clin Gastroenterol
disease supports the presence of multiple fibrogenic Hepatol 2019;17:616–629.e26.
pathways. Liver Int 2013;33:624–632. 28. McPherson S, Hardy T, Henderson E, et al. Evidence of
12. Dufour JF. Time to Abandon NASH? Hepatology 2016; NAFLD progression from steatosis to fibrosing-
63:9–10. steatohepatitis using paired biopsies: implications for
13. Bellentani S, Tiribelli C. Is it time to change NAFLD and prognosis and clinical management. J Hepatol 2015;
NASH nomenclature? Lancet Gastroenterol Hepatol 62:1148–1155.
2017;2:547–548. 29. Nobili V, Alisi A, Valenti L, et al. NAFLD in children: new
14. Loria P, Lonardo A, Carulli N. Should nonalcoholic fatty genes, new diagnostic modalities and new drugs. Nat
liver disease be renamed? Dig Dis 2005;23:72–82. Rev Gastroenterol Hepatol 2019;16:517–530.
15. Eslam M, Sanyal AJ, George J. Toward more accurate 30. Mardinoglu A, Uhlen M, Boren J. Broad Views of non-
nomenclature for fatty liver diseases. Gastroenterology alcoholic fatty liver disease. Cell Syst 2018;6:7–9.
2019;157:590–593. 31. Pirola CJ, Sookoian S. Multiomics biomarkers for the
16. Dalkey NC. Studies in the Quality of Life; Delphi and prediction of nonalcoholic fatty liver disease severity.
Decision-Making. Lexington, MA: Lexington Books; World J Gastroenterol 2018;24:1601–1615.
1972. 32. Ong JP, Pitts A, Younossi ZM. Increased overall mor-
17. Yki-Jarvinen H, Luukkonen PK. Heterogeneity of non- tality and liver-related mortality in non-alcoholic fatty
alcoholic fatty liver disease. Liver Int 2015;35:2498– liver disease. J Hepatol 2008;49:608–612.
2500. 33. Frith J, Day CP, Henderson E, et al. Non-alcoholic fatty
18. Lonardo A, Ballestri S, Targher G. “Not all forms of liver disease in older people. Gerontology 2009;55:607–
NAFLD were created equal.” Do metabolic syndrome- 613.
related NAFLD and PNPLA3-related NAFLD exert a 34. Frith J, Jones D, Newton JL. Chronic liver disease in an
variable impact on the risk of early carotid atheroscle- ageing population. Age Ageing 2009;38:11–18.
rosis? Atherosclerosis 2017;257:253–255. 35. Donati B, Telomeres VL. NAFLD and chronic liver dis-
19. Adams LA, Lymp JF, St Sauver J, et al. The natural ease. Int J Mol Sci 2016;17:383.
history of nonalcoholic fatty liver disease: a population- 36. Papatheodoridi AM, Chrysavgis L, Koutsilieris M, et al.
based cohort study. Gastroenterology 2005;129:113– The role of senescence in the development of non-
121. alcoholic fatty liver disease and progression to non-
20. Angulo P, Kleiner DE, Dam-Larsen S, et al. Liver fibrosis, alcoholic steatohepatitis. Hepatology 2020;71:363–374.
but no other histologic features, is associated with long- 37. Kuk JL, Saunders TJ, Davidson LE, et al. Age-related
term outcomes of patients with nonalcoholic fatty liver changes in total and regional fat distribution. Ageing Res
disease. Gastroenterology 2015;149:389–397.e10. Rev 2009;8:339–348.
May 2020 MAFLD: A Proposed Nomenclature 2009

38. Churilla JR, Fitzhugh EC, Thompson DL. The metabolic 54. Zhou F, Zhou J, Wang W, et al. Unexpected rapid in-
syndrome: how definition impacts the prevalence and crease in the burden of nonalcoholic fatty liver disease in
risk in US adults: 1999–2004 NHANES. Metab Syndr China From 2008 to 2018: a systematic review and
Relat Disord 2007;5:331–342. meta-analysis. Hepatology 2019;70:1119–1133.
39. Lee YH, Jung KS, Kim SU, et al. Sarcopaenia is asso- 55. Weston SR, Leyden W, Murphy R, et al. Racial and
ciated with NAFLD independently of obesity and insulin ethnic distribution of nonalcoholic fatty liver in persons
resistance: Nationwide surveys (KNHANES 2008–2011). with newly diagnosed chronic liver disease. Hepatology
J Hepatol 2015;63:486–493. 2005;41:372–379.
40. Lee YH, Kim SU, Song K, et al. Sarcopenia is associated 56. Mohanty SR, Troy TN, Huo D, et al. Influence of ethnicity
with significant liver fibrosis independently of obesity on histological differences in non-alcoholic fatty liver
and insulin resistance in nonalcoholic fatty liver disease: disease. J Hepatol 2009;50:797–804.
nationwide surveys (KNHANES 2008–2011). Hepatology 57. Tabibian JH, Lazo M, Durazo FA, et al. Nonalcoholic fatty
2016;63:776–786. liver disease across ethno-racial groups: do Asian-
41. Lonardo A, Nascimbeni F, Ballestri S, et al. Sex differ- American adults represent a new at-risk population?
ences in NAFLD: state of the art and identification of J Gastroenterol Hepatol 2011;26:501–509.
research gaps. Hepatology 2019;70:1457–1469. 58. Wong VWS, Chu WCW, Wong GLH, et al. Prevalence of
42. Anderson EL, Howe LD, Jones HE, et al. The prevalence non-alcoholic fatty liver disease and advanced fibrosis in
of non-alcoholic fatty liver disease in children and ado- Hong Kong Chinese: a population study using proton-
lescents: a systematic review and meta-analysis. PLoS magnetic resonance spectroscopy and transient elas-
One 2015;10:e0140908. tography. Gut 2012;61:409–415.
43. Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver 59. Browning JD, Szczepaniak LS, Dobbins R, et al. Preva-
disease. Gastroenterology 2002;122:1649–1657. lence of hepatic steatosis in an urban population in the
44. Yang JD, Abdelmalek MF, Pang H, et al. Gender and United States: impact of ethnicity. Hepatology 2004;
menopause impact severity of fibrosis among patients 40:1387–1395.
with nonalcoholic steatohepatitis. Hepatology 2014; 60. Guerrero R, Vega GL, Grundy SM, et al. Ethnic differ-
59:1406–1414. ences in hepatic steatosis: an insulin resistance
45. Yang JD, Suzuki A. The influence of menopause on the paradox? Hepatology 2009;49:791–801.
development of hepatic fibrosis in nonobese women 61. Romeo S, Kozlitina J, Xing C, et al. Genetic variation in
with nonalcoholic fatty liver disease: reply. Hepatology PNPLA3 confers susceptibility to nonalcoholic fatty liver
2014;60:1792–1793. disease. Nat Genet 2008;40:1461–1465.
46. Klair JS, Yang JD, Abdelmalek MF, et al. A longer 62. Shen J, Wong GLH, Chan HLY, et al. PNPLA3 gene
duration of estrogen deficiency increases fibrosis risk polymorphism accounts for fatty liver in community
among postmenopausal women with nonalcoholic fatty subjects without metabolic syndrome. Aliment Pharma-
liver disease. Hepatology 2016;64:85–91. col Ther 2014;39:532–539.
47. Norheim F, Hui ST, Kulahcioglu E, et al. Genetic and 63. Guthold R, Stevens GA, Riley LM, et al. Worldwide trends
hormonal control of hepatic steatosis in female and male in insufficient physical activity from 2001 to 2016: a pooled
mice. J Lipid Res 2017;58:178–187. analysis of 358 population-based surveys with 1.9 million
48. Ganz M, Csak T, Szabo G. High fat diet feeding results in participants. Lancet Glob Health 2018;6:e1077–e1086.
gender specific steatohepatitis and inflammasome acti- 64. Bambha K, Belt P, Abraham M, et al. Ethnicity and nonal-
vation. World J Gastroenterol 2014;20:8525–8534. coholic fatty liver disease. Hepatology 2012;55:769–780.
49. Lovejoy JC, Champagne CM, de Jonge L, et al. 65. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis
Increased visceral fat and decreased energy expenditure and management of nonalcoholic fatty liver disease:
during the menopausal transition. Int J Obes (Lond) practice guidance from the American Association for the
2008;32:949–958. Study of Liver Diseases. Hepatology 2018;67:328–357.
50. Kurt Z, Barrere-Cain R, LaGuardia J, et al. Tissue-spe- 66. Saad L. Majority in US drink alcohol, averaging four
cific pathways and networks underlying sexual dimor- drinks a week. Gallup Well-Being August 17, 2012.
phism in non-alcoholic fatty liver disease. Biol Sex Differ Available at: https://news.gallup.com/poll/156770/
2018;9:46. majority-drink-alcohol-averaging-four-drinks-week.aspx.
51. Tomas TC, Urlep Z, Moskon M, et al. LiverSex compu- Accessed April 9, 2020.
tational model: sexual aspects in hepatic metabolism 67. Dunn W, Sanyal AJ, Brunt EM, et al. Modest alcohol
and abnormalities. Front Physiol 2018;9:360. consumption is associated with decreased prevalence of
52. Marin V, Rosso N, Dal Ben M, et al. An animal model for steatohepatitis in patients with non-alcoholic fatty liver
the juvenile non-alcoholic fatty liver disease and non- disease (NAFLD). J Hepatol 2012;57:384–391.
alcoholic steatohepatitis. PLoS One 2016;11: 68. Kwon HK, Greenson JK, Conjeevaram HS. Effect of lifetime
e0158817. alcohol consumption on the histological severity of non-
53. Rich NE, Oji S, Mufti AR, et al. Racial and ethnic dis- alcoholic fatty liver disease. Liver Int 2014;34:129–135.
parities in nonalcoholic fatty liver disease prevalence, 69. Ekstedt M, Franzen LE, Holmqvist M, et al. Alcohol
severity, and outcomes in the United States: a system- consumption is associated with progression of hepatic
atic review and meta-analysis. Clin Gastroenterol Hep- fibrosis in non-alcoholic fatty liver disease. Scand J
atol 2018;16:198–210.e2. Gastroenterol 2009;44:366–374.
2010 Eslam et al Gastroenterology Vol. 158, No. 7

70. Ascha MS, Hanouneh IA, Lopez R, et al. The incidence 85. Leung C, Rivera L, Furness JB, et al. The role of the gut
and risk factors of hepatocellular carcinoma in patients microbiota in NAFLD. Nat Rev Gastroenterol Hepatol
with nonalcoholic steatohepatitis. Hepatology 2010; 2016;13:412–425.
51:1972–1978. 86. Hoyles L, Fernandez-Real JM, Federici M, et al. Molec-
71. Meltzer-Brody S, Colquhoun H, Riesenberg R. GBD ular phenomics and metagenomics of hepatic steatosis
2016 Alcohol Collaborators. Alcohol use and burden in non-diabetic obese women. Nat Med 2018;24:1070–
for 195 countries and territories, 1990–2016: a sys- 1080.
tematic analysis for the Global Burden of Disease 87. Loomba R, Seguritan V, Li W, et al. Gut microbiome-
Study 2016 (vol 392, pg 1015, 2018). Lancet 2018; based metagenomic signature for non-invasive detec-
392:1116. tion of advanced fibrosis in human nonalcoholic fatty
72. VanWagner LB, Ning HY, Allen NB, et al. Alcohol use and liver disease. Cell Metab 2017;25:1054–1062.e5.
cardiovascular disease risk in patients with nonalcoholic 88. Caussy C, Tripathi A, Humphrey G, et al. A gut micro-
fatty liver disease. Gastroenterology 2017;153:1260– biome signature for cirrhosis due to nonalcoholic fatty
1272.e3. liver disease. Nat Commun 2019;10:1406.
73. Boyle M, Masson S, Anstee QM. The bidirectional im- 89. Caussy C, Hsu C, Lo MT, et al. Link between gut-
pacts of alcohol consumption and the metabolic syn- microbiome derived metabolite and shared gene-
drome: cofactors for progressive fatty liver disease. effects with hepatic steatosis and fibrosis in NAFLD.
J Hepatol 2018;68:251–267. Hepatology 2018;68:918–932.
74. Aberg F, Helenius-Hietala J, Puukka P, et al. Interaction 90. Caussy C, Loomba R. Gut microbiome, microbial me-
between alcohol consumption and metabolic syndrome tabolites and the development of NAFLD. Nat Rev
in predicting severe liver disease in the general popula- Gastroenterol Hepatol 2018;15:719–720.
tion. Hepatology 2018;67:2141–2149. 91. Arab JP, Karpen SJ, Dawson PA, et al. Bile acids and
75. Chang Y, Cho YK, Kim Y, et al. Nonheavy drinking and nonalcoholic fatty liver disease: Molecular insights
worsening of noninvasive fibrosis markers in nonalco- and therapeutic perspectives. Hepatology 2017;65:350–
holic fatty liver disease: a cohort study. Hepatology 362.
2019;69:64–75. 92. Ilan Y. Leaky gut and the liver: a role for bacterial
76. Younossi ZM, Stepanova M, Ong J, et al. Effects of translocation in nonalcoholic steatohepatitis. World J
alcohol consumption and metabolic syndrome on mor- Gastroenterol 2012;18:2609–2618.
tality in patients with non-alcoholic and alcohol-related 93. Enomoto N, Ikejima K, Yamashina S, et al. Kupffer cell
fatty liver disease. Clin Gastroenterol Hepatol 2018; sensitization by alcohol involves increased permeability
17:1625–1633.e1. to gut-derived endotoxin. Alcohol Clin Exp Res 2001;
77. Åberg F, Puukka P, Salomaa V, et al. Risks of light and 25:51S–54S.
moderate alcohol use in fatty liver disease–follow-up of 94. Miura K, Yang L, van Rooijen N, et al. Toll-like receptor 2
population cohorts [published online ahead of print July and palmitic acid cooperatively contribute to the
19, 2019]. Hepatology https://doi.org/10.1002/hep. development of nonalcoholic steatohepatitis through
30864. inflammasome activation in mice. Hepatology 2013;
78. Ajmera V, Belt P, Wilson LA, et al. Among patients with 57:577–589.
nonalcoholic fatty liver disease, modest alcohol use is 95. Kawai T, Akira S. TLR signaling. Semin Immunol 2007;
associated with less improvement in histologic steatosis 19:24–32.
and steatohepatitis. Clin Gastroenterol Hepatol 2018; 96. Jialal I, Kaur H, Devaraj S. Toll-like receptor status in
16:1511–1520.e5. obesity and metabolic syndrome: a translational
79. Lackner C, Tiniakos D. Fibrosis and alcohol-related liver perspective. J Clin Endocrinol Metab 2014;99:39–48.
disease. J Hepatol 2019;70:294–304. 97. Wang J, Thingholm LB, Skieceviciene J, et al. Genome-
80. Rehm J, Taylor B, Mohapatra S, et al. Alcohol as a risk wide association analysis identifies variation in vitamin D
factor for liver cirrhosis: a systematic review and meta- receptor and other host factors influencing the gut
analysis. Drug Alcohol Rev 2010;29:437–445. microbiota. Nat Genet 2016;48:1396–1406.
81. Romero-Gomez M, Zelber-Sagi S, Trenell M. Treatment 98. Bonder MJ, Kurilshikov A, Tigchelaar EF, et al. The effect
of NAFLD with diet, physical activity and exercise. of host genetics on the gut microbiome. Nat Genet 2016;
J Hepatol 2017;67:829–846. 48:1407–1412.
82. Trovato FM, Catalano D, Martines GF, et al. Mediterra- 99. Hall AB, Tolonen AC, Xavier RJ. Human genetic variation
nean diet and non-alcoholic fatty liver disease. The need and the gut microbiome in disease. Nat Rev Genet 2017;
of extended and comprehensive interventions. Clin Nutr 18:690–699.
2015;34:86–88. 100. Brooks AW, Priya S, Blekhman R, et al. Gut microbiota
83. Estruch R, Ros E, Salas-Salvado J, et al. Primary pre- diversity across ethnicities in the United States. PLoS
vention of cardiovascular disease with a mediterranean Biology 2018;16:e2006842.
diet supplemented with extra-virgin olive oil or nuts. 101. Despres JP. Body fat distribution and risk of cardio-
N Engl J Med 2018;378:e34. vascular disease: an update. Circulation 2012;126:1301–
84. David LA, Maurice CF, Carmody RN, et al. Diet rapidly 1313.
and reproducibly alters the human gut microbiome. 102. Loos RJF, Kilpelainen TO. Genes that make you fat, but
Nature 2014;505:559–563. keep you healthy. J Intern Med 2018;284:450–463.
May 2020 MAFLD: A Proposed Nomenclature 2011

103. Chitturi S, George J. Interaction of iron, insulin resis- 119. Fehlert E, Wagner R, Ketterer C, et al. Genetic determi-
tance, and nonalcoholic steatohepatitis. Curr Gastro- nation of body fat distribution and the attributive influ-
enterol Rep 2003;5:18–25. ence on metabolism. Obesity 2017;25:1277–1283.
104. Eckel N, Li Y, Kuxhaus O, et al. Transition from metabolic 120. Lotta LA, Gulati P, Day FR, et al. Integrative genomic
healthy to unhealthy phenotypes and association with analysis implicates limited peripheral adipose storage
cardiovascular disease risk across BMI categories in 90 capacity in the pathogenesis of human insulin resistance
257 women (the Nurses’ Health Study): 30 year follow- (vol 49, pg 17, 2016). Nat Genet 2017;49:317–317.
up from a prospective cohort study. Lancet Diabetes 121. Ji Y, Yiorkas AM, Frau F, et al. Genome-wide and
Endocrinol 2018;6:714–724. abdominal MRI-imaging data provides evidence that a
105. Caleyachetty R, Thomas GN, Toulis KA, et al. Metabol- genetically determined favourable adiposity phenotype
ically healthy obese and incident cardiovascular disease is characterized by lower ectopic liver fat and lower risk
events among 3.5 million men and women. J Am Coll of type 2 diabetes, heart disease and hypertension.
Cardiol 2017;70:1429–1437. Diabetes 2019;68:207–219.
106. Lassale C, Tzoulaki I, Moons KGM, et al. Separate and 122. Yaghootkar H, Lotta L, Tyrrell J, et al. Genetic evidence
combined associations of obesity and metabolic health for a link between favorable adiposity and lower risk of
with coronary heart disease: a pan-European case- type 2 diabetes, hypertension, and heart disease. Dia-
cohort analysis. Eur Heart J 2018;39:397–406. betes 2016;65:A421.
107. Winkler TW, Gunther F, Hollerer S, et al. A joint view on 123. Luukkonen PK, Zhou Y, Sädevirta S, et al. Hepatic
genetic variants for adiposity differentiates subtypes ceramides dissociate steatosis and insulin resistance in
with distinct metabolic implications (vol 9, 2018). Nat patients with non-alcoholic fatty liver disease. J Hepatol
Commun 2018;9:1946. 2016;64:1167–1175.
108. van der Poorten D, Milner KL, Hui J, et al. Visceral fat: a 124. Alonso C, Fernandez-Ramos D, Varela-Rey M, et al.
key mediator of steatohepatitis in metabolic liver dis- Metabolomic identification of subtypes of nonalcoholic
ease. Hepatology 2008;48:449–457. steatohepatitis. Gastroenterology 2017;152:1449–1461.
109. Perseghin G. Lipids in the wrong place: visceral fat e7.
and nonalcoholic steatohepatitis. Diabetes Care 2011; 125. Scupakova K, Soons Z, Ertaylan G, et al. Spatial systems
34(Suppl 2):S367–S370. lipidomics reveals nonalcoholic fatty liver disease het-
110. Agbim U, Carr RM, Pickett-Blakely O, et al. Ethnic dis- erogeneity. Analyt Chem 2018;90:5130–5138.
parities in adiposity: focus on non-alcoholic fatty liver 126. Hoang S, Oseini AM, Vincent RK, et al. Development and
disease, visceral, and generalized obesity. Curr Obes validation of a gene-level molecular disease activity and
Rep 2019;8:243–254. fibrosis score for nonalcoholic fatty liver disease.
111. Lim U, Ernst T, Buchthal SD, et al. Asian women have Hepatology 2018;68(1):SUPPL abs.1286.
greater abdominal and visceral adiposity than Caucasian 127. Das K, Chowdhury A. Lean NASH: distinctiveness
women with similar body mass index. Nutr Diabetes and clinical implication. Hepatol Int 2013;7(Suppl 2):806–
2011;1:e6. 813.
112. Araneta MR, Barrett-Connor E. Ethnic differences in 128. Ding C, Chan ZL, Magkos F. Lean, but not healthy: the
visceral adipose tissue and type 2 diabetes: Filipino, “metabolically obese, normal-weight” phenotype. Curr
African-American, and white women. Obes Res 2005; Opin Clin Nutr Metabolic Care 2016;19:408–417.
13:1458–1465. 129. Dela Cruz AC, Bugianesi E, George J, et al. Character-
113. Lear SA, Humphries KH, Kohli S, et al. The use of BMI istics and long-term prognosis of lean patients with
and waist circumference as surrogates of body fat differs nonalcoholic fatty liver disease. Gastroenterology 2014;
by ethnicity. Obesity 2007;15:2817–2824. 146:S909.
114. Park YW, Allison DB, Heymsfield SB, et al. Larger 130. Hagstrom H, Nasr P, Ekstedt M, et al. Risk for devel-
amounts of visceral adipose tissue in Asian Americans. opment of severe liver disease in lean patients with
Obes Res 2001;9:381–387. nonalcoholic fatty liver disease: a long-term follow-up
115. Chandalia M, Lin P, Seenivasan T, et al. Insulin resis- study. Hepatol Commun 2018;2:48–57.
tance and body fat distribution in South Asian men 131. Leung JCF, Loong TCW, Wei JL, et al. Histological
compared to Caucasian men. PLoS One 2007;2:e812. severity and clinical outcomes of nonalcoholic fatty liver
116. Petersen KF, Dufour S, Feng J, et al. Increased preva- disease in nonobese patients. Hepatology 2017;65:54–
lence of insulin resistance and nonalcoholic fatty liver 64.
disease in Asian-Indian men. Proc Natl Acad Sci U S A 132. Wei JL, Leung JCF, Loong TCW, et al. Prevalence and
2006;103:18273–18277. severity of nonalcoholic fatty liver disease in non-obese
117. Gujral UP, Pradeepa R, Weber MB, et al. Type 2 diabetes patients: a population study using proton-magnetic
in South Asians: similarities and differences with white resonance spectroscopy. Am J Gastroenterol 2015;
Caucasian and other populations. Ann N Y Acad Sci 110:1306–1314.
2013;1281:51–63. 133. Chen F, Esmaili S, Rogers G, et al. Lean NAFLD: a
118. Shungin D, Winkler TW, Croteau-Chonka DC, et al. New distinct entity shaped by differential metabolic adapta-
genetic loci link adipose and insulin biology to body fat tion [published online ahead of print August 23, 2019].
distribution. Nature 2015;518:187–196. Hepatology https://doi.org/10.1002/hep.30908.
2012 Eslam et al Gastroenterology Vol. 158, No. 7

134. Fracanzani AL, Petta S, Lombardi R, et al. Liver and 151. Holmen OL, Zhang H, Fan YB, et al. Systematic evalu-
cardiovascular damage in patients with lean nonalco- ation of coding variation identifies a candidate
holic fatty liver disease, and association with visceral causal variant in TM6SF2 influencing total cholesterol
obesity. Clin Gastroenterol Hepatol 2017;15:1604–1611. and myocardial infarction risk. Nat Genet 2014;46:345–
135. Loomba R, Schork N, Chen CH, et al. Heritability of 351.
hepatic fibrosis and steatosis based on a prospective 152. Kozlitina J, Smagris E, Stender S, et al. Exome-wide
twin study. Gastroenterology 2015;149:1784–1793. association study identifies a TM6SF2 variant that con-
136. Struben VM, Hespenheide EE, Caldwell SH. Nonalco- fers susceptibility to nonalcoholic fatty liver disease. Nat
holic steatohepatitis and cryptogenic cirrhosis within Genet 2014;46:352–356.
kindreds. Am J Med 2000;108:9–13. 153. Liu YL, Reeves HL, Burt AD, et al. TM6SF2 rs58542926
137. Caussy C, Soni M, Cui J, et al. Nonalcoholic fatty liver influences hepatic fibrosis progression in patients with
disease with cirrhosis increases familial risk for non-alcoholic fatty liver disease. Nat Commun 2014;
advanced fibrosis. J Clin Invest 2017;127:2697–2704. 5:4309.
138. Eslam M, George J. Genetic and epigenetic mechanisms 154. Eslam M, Mangia A, Berg T, et al. Diverse impacts of the
of NASH. Hepatol Int 2016;10:394–406. rs58542926 E167K variant in TM6SF2 on viral and
139. Eslam M, Valenti L, Romeo S. Genetics and epigenetics metabolic liver disease phenotypes. Hepatology 2016;
of NAFLD and NASH: clinical impact. J Hepatol 2018; 64:34–46.
68:268–279. 155. Speliotes EK, Yerges-Armstrong LM, Wu J, et al.
140. Mohlenberg M, Terczynska-Dyla E, Thomsen KL, et al. Genome-wide association analysis identifies variants
The role of IFN in the development of NAFLD and NASH. associated with nonalcoholic fatty liver disease that have
Cytokine 2018;124:154519. distinct effects on metabolic traits. PLoS Genetics 2011;
141. Eslam M, Hashem AM, Leung R, et al. Interferon-lambda 7:e1001324.
rs12979860 genotype and liver fibrosis in viral and 156. Liu DJ, Peloso GM, Yu H, et al. Exome-wide association
non-viral chronic liver disease. Nat Commun 2015; study of plasma lipids in > 300,000 individuals. Nat
6:6422. Genet 2017;49:1758–1766.
142. Eslam M, McLeod D, Kelaeng KS, et al. IFN-lambda 3, 157. Diogo D, Tian C, Franklin CS, et al. Phenome-wide as-
not IFN-lambda 4, likely mediates IFNL3-IFNL4 sociation studies across large population cohorts sup-
haplotype-dependent hepatic inflammation and fibrosis. port drug target validation. Nat Commun 2018;9:4285.
Nat Genet 2017;49:795–800. 158. Mahajan A, Wessel J, Willems SM, et al. Refining the
143. Metwally M, Bayoumi A, Romero-Gomez M, et al. accuracy of validated target identification through cod-
A polymorphism in the Irisin-encoding gene (FNDC5) ing variant fine-mapping in type 2 diabetes. Nat Genet
associates with hepatic steatosis by differential miRNA 2018;50:559–571.
binding to the 3’UTR. J Hepatol 2019;70:494–500. 159. Abul-Husn NS, Cheng X, Li AH, et al. A protein-trun-
144. Sookoian S, Pirola CI. Genetics of nonalcoholic fatty liver cating HSD17B13 variant and protection from chronic
disease: from pathogenesis to therapeutics. Semin Liver liver disease. N Engl J Med 2018;378:1096–1106.
Dis 2019;39:124–140. 160. Mancina RM, Dongiovanni P, Petta S, et al. The
145. Sookoian S, Pirola CJ. Genetic predisposition in nonal- MBOAT7-TMC4 variant rs641738 increases risk of
coholic fatty liver disease. Clin Molec Hepatol 2017; nonalcoholic fatty liver disease in individuals of European
23:1–12. descent. Gastroenterology 2016;150:1219–1230.e6.
146. Pelusi S, Baselli G, Pietrelli A, et al. Rare pathogenic 161. Simons N, Isaacs A, Koek GH, et al. PNPLA3, TM6SF2,
variants predispose to hepatocellular carcinoma in and MBOAT7 genotypes and coronary artery disease.
nonalcoholic fatty liver disease. Sci Rep 2019;9:3682. Gastroenterology 2017;152:912–913.
147. Nobili V, Liccardo D, Bedogni G, et al. Influence of di- 162. Thabet K, Asimakopoulos A, Shojaei M, et al. MBOAT7
etary pattern, physical activity, and I148M PNPLA3 on rs641738 increases risk of liver inflammation and tran-
steatosis severity in at-risk adolescents. Genes Nutr sition to fibrosis in chronic hepatitis C. Nat Commun
2014;9:392. 2016;7:12757.
148. Davis JN, Le KA, Walker RW, et al. Increased hepatic fat 163. Liu CH, Ampuero J, Gil-Gomez A, et al. miRNAs in pa-
in overweight Hispanic youth influenced by interaction tients with non-alcoholic fatty liver disease: A systematic
between genetic variation in PNPLA3 and high dietary review and meta-analysis. J Hepatol 2018;69:1335–1348.
carbohydrate and sugar consumption. Am J Clin Nutr 164. Pirola CJ, Gianotti TF, Castano GO, et al. Circulating
2010;92:1522–1527. microRNA signature in non-alcoholic fatty liver disease:
149. Santoro N, Savoye M, Kim G, et al. Hepatic fat accu- from serum non-coding RNAs to liver histology and
mulation is modulated by the interaction between the disease pathogenesis. Gut 2015;64:800–812.
rs738409 variant in the PNPLA3 gene and the dietary 165. Atanasovska B, Rensen SS, van der Sijde MR, et al.
Omega6/Omega3 PUFA intake. PLoS One 2012;7: A liver-specific long noncoding RNA with a role in cell
e37827. viability is elevated in human nonalcoholic steatohepa-
150. Barata L, Feitosa MF, Bielak LF, et al. Insulin resistance titis. Hepatology 2017;66:794–808.
exacerbates genetic predisposition to nonalcoholic fatty 166. Zhao XY, Xiong XL, Liu TY, et al. Long noncoding RNA
liver disease in individuals without diabetes. Hepatol licensing of obesity-linked hepatic lipogenesis and
Commun 2019;3:894–907. NAFLD pathogenesis. Nat Commun 2018;9:2986.
May 2020 MAFLD: A Proposed Nomenclature 2013

167. Sookoian S, Flichman D, Garaycoechea ME, et al. diabetes in mammals. Proc Natl Acad Sci U S A 2014;
Metastasis-associated lung adenocarcinoma transcript 111:1873–1878.
1 as a common molecular driver in the pathogenesis of 183. Aiken CE, Tarry-Adkins JL, Ozanne SE. Transgenera-
nonalcoholic steatohepatitis and chronic immune medi- tional developmental programming of ovarian reserve.
ated liver damage. Hepatol Commun 2018;2:654–665. Sci Rep 2015;5:16175.
168. Sookoian S, Rohr C, Salatino A, et al. Genetic variation in 184. Laker RC, Connelly JJ, Yan Z. Exercise prevents
long noncoding RNAs and the risk of nonalcoholic fatty maternal high-fat diet-induced hypermethylation of the
liver disease. Oncotarget 2017;8:22917–22926. Pgc-1a gene and age-dependent metabolic dysfunction
169. Ahrens M, Ammerpohl O, von Schonfels W, et al. DNA in the offspring. Diabetes 2014;63:160521611.
methylation analysis in nonalcoholic fatty liver disease 185. Barres R, Zierath JR. The role of diet and exercise in the
suggests distinct disease-specific and remodeling sig- transgenerational epigenetic landscape of T2DM. Nat
natures after bariatric surgery. Cell Metab 2013;18:296– Rev Endocrinol 2016;12:441–451.
302. 186. Ajmera VH, Terrault NA, VanWagner LB, et al. Longer
170. Murphy SK, Yang HN, Moylan CA, et al. Relationship lactation duration is associated with decreased preva-
between methylome and transcriptome in patients with lence of non-alcoholic fatty liver disease in women.
nonalcoholic fatty liver disease. Gastroenterology 2013; J Hepatol 2019;70:126–132.
145:1076–1087. 187. Ayonrinde OT, Oddy WH, Adams LA, et al. Infant nutri-
171. Ma J, Nano J, Ding J, et al. A peripheral blood DNA tion and maternal obesity influence the risk of non-
methylation signature of hepatic fat reveals a potential alcoholic fatty liver disease in adolescents. J Hepatol
causal pathway for non-alcoholic fatty liver disease. 2017;67:568–576.
Diabetes 2019;68:1073–1083. 188. Sookoian S, Rosselli MS, Gemma C, et al. Epigenetic
172. Hardy T, Zeybel M, Day CP, et al. Plasma DNA methyl- regulation of insulin resistance in nonalcoholic fatty
ation: a potential biomarker for stratification of liver liver disease: impact of liver methylation of the
fibrosis in non-alcoholic fatty liver disease. Gut 2017; peroxisome proliferator-activated receptor gamma
66:1321–1328. coactivator 1alpha promoter. Hepatology 2010;52:1992–
173. Kitamoto T, Kitamoto A, Ogawa Y, et al. Targeted- 2000.
bisulfite sequence analysis of the methylation of CpG 189. Gemma C, Sookoian S, Alvarinas J, et al. Maternal
islands in genes encoding PNPLA3, SAMM50, and pregestational BMI is associated with methylation of the
PARVB of patients with non-alcoholic fatty liver disease. PPARGC1A promoter in newborns. Obesity 2009;
J Hepatol 2015;63:494–502. 17:1032–1039.
174. De Chiara F, Heeboll S, Marrone G, et al. Urea cycle 190. McPherson S, Hardy T, Dufour JF, et al. Age as a con-
dysregulation in non-alcoholic fatty liver disease. founding factor for the accurate non-invasive diagnosis
J Hepatol 2018;69:905–915. of advanced NAFLD fibrosis. Am J Gastroenterol 2017;
175. Jalan R, De Chiara F, Balasubramaniyan V, et al. 112:740–751.
Ammonia produces pathological changes in human he- 191. De Silva S, Li WH, Kemos P, et al. Non-invasive markers
patic stellate cells and is a target for therapy of portal of liver fibrosis in fatty liver disease are unreliable in
hypertension. J Hepatol 2016;64:823–833. people of South Asian descent. Frontline Gastroenterol
176. Thomsen KL, De Chiara F, Rombouts K, et al. Ammonia: 2018;9:115–121.
a novel target for the treatment of non-alcoholic stea- 192. Santhekadur PK, Kumar DP, Sanyal AJ. Preclinical
tohepatitis. Med Hypotheses 2018;113:91–97. models of non-alcoholic fatty liver disease. J Hepatol
177. De Chiara F, Thomsen KL, Habtesion A, et al. Ammonia 2018;68:230–237.
scavenging prevents progression of fibrosis in experi- 193. Tu LN, Showalter MR, Cajka T, et al. Metabolomic
mental non-alcoholic fatty liver disease. Hepatology characteristics of cholesterol-induced non-obese
2019. nonalcoholic fatty liver disease in mice. Sci Rep 2017;
178. Baker PR 2nd, Friedman JE. Mitochondrial role in the 7:6120.
neonatal predisposition to developing nonalcoholic fatty 194. Drew L. Development pipeline review 2018. Nature 2017;
liver disease. J Clin Invest 2018;128:3692–3703. 551:S86–S89.
179. Li Y. Epigenetic mechanisms link maternal diets and gut 195. Woodcock J, LaVange LM. Master protocols to study
microbiome to obesity in the offspring. Front Genet multiple therapies, multiple diseases, or both. N Engl J
2018;9:342. Med 2017;377:62–70.
180. Suter MA, Ma J, Vuguin PM, et al. In utero exposure to a 196. Bhatt DL, Mehta C. Adaptive designs for clinical trials.
maternal high-fat diet alters the epigenetic histone code N Engl J Med 2016;375:65–74.
in a murine model. Am J Obstet Gynecol 2014;210:463. 197. Brunt EM, Ramrakhiani S, Cordes BG, et al. Concur-
e1–463.e11. rence of histologic features of steatohepatitis with other
181. Suter MA, Chen A, Burdine MS, et al. A maternal high-fat forms of chronic liver disease. Mod Pathol 2003;16:49–
diet modulates fetal SIRT1 histone and protein deace- 56.
tylase activity in nonhuman primates. FASEB J 2012; 198. Sanchez-Munoz D, Castellano-Megias VM, Romero-
26:5106–5114. Gomez M. Histologic features of steatohepatitis in
182. Wei YC, Yang CR, Wei YP, et al. Paternally induced patients with a clinical diagnosis of autoimmune chole-
transgenerational inheritance of susceptibility to stasis. Dig Dis Sci 2004;49:1957–1961.
2014 Eslam et al Gastroenterology Vol. 158, No. 7

199. Cotrim HP, Andrade ZA, Parana R, et al. Nonalcoholic Mary Rinella, Department of Gastroenterology, Northwestern University
Feinberg School of Medicine, Chicago, Illinois.
steatohepatitis: a toxic liver disease in industrial workers. Marco Arrese, Departamento de Gastroenterología, Escuela de Medicina,
Liver 1999;19:299–304. Pontificia Universidad Católica de Chile, Santiago, Chile.
200. Choi HS, Brouwer WP, Zanjir WM, et al. Non-alcoholic Mohammed Eslam, Storr Liver Centre, Westmead Institute for Medical
Research, Westmead Hospital and University of Sydney, NSW, Australia.
steatohepatitis is associated with liver-related outcomes Pierre Bedossa, Department of Pathology, Physiology and Imaging, Beaujon
and all-cause mortality in chronic hepatitis B. Hepatol- Hospital Paris Diderot University, Paris, France.
Philip N. Newsome, National Institute for Health Research Biomedical
ogy 2020;71:539–548. Research Centre at University Hospitals Birmingham NHS Foundation Trust
201. Chiang DJ, McCullough AJ. The impact of obesity and and the University of Birmingham, UK; Centre for Liver and Gastrointestinal
metabolic syndrome on alcoholic liver disease. Clin Liver Research, Institute of Immunology and Immunotherapy, University of
Birmingham, UK; Liver Unit, University Hospitals Birmingham NHS
Dis 2014;18:157–163. Foundation Trust, Birmingham, UK.
202. Beuers U, Gershwin ME, Gish RG, et al. Changing Quentin M. Anstee, Institute of Cellular Medicine, Faculty of Medical
Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom;
nomenclature for PBC: from ‘cirrhosis’ to ‘cholangitis’. Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne
Gastroenterology 2015;149:1627–1629. Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
Rajiv Jalan, Liver Failure Group, Institute for Liver and Digestive Health,
University College London, Royal Free Campus, London, United Kingdom.
Ramon Bataller, Division of Gastroenterology, Hepatology and Nutrition,
Received September 10, 2019. Accepted November 5, 2019. University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
Rohit Loomba, Division of Epidemiology, Department of Family Medicine and
Correspondence Public Health, University of California at San Diego, La Jolla, California.
Address correspondence to: Mohammed Eslam, Storr Liver Centre, Westmead Silvia Sookoian, Department of Clinical and Molecular Hepatology, National
Institute for Medical Research, Westmead Hospital and University of Sydney, Scientific and Technical Research Council (CONICET), University of Buenos
Westmead 2145, NSW, Australia. e-mail: mohammed.eslam@sydney.edu.au; Aires, Institute of Medical Research (IDIM), Ciudad Autónoma de Buenos
fax: +61-2-96357582; or Arun J. Sanyal, Virginia Commonwealth University Aires, Argentina.
School of Medicine, MCV Box 980341, Richmond, Virginia 23298-0341. Shiv K. Sarin, Department of Hepatology, Institute of Liver and Biliary
e-mail: arun.sanyal@vcuhealth.org; fax: +1 (804) 828 2992; or Jacob Sciences, New Delhi, India.
George, MD, Storr Liver Centre, Westmead Institute for Medical Research, Stephen Harrison, Radcliffe Department of Medicine, University of Oxford,
Westmead Hospital and University of Sydney, Westmead 2145, NSW, Oxford, UK.
Australia. e-mail: jacob.george@sydney.edu.au; fax: +61-2-96357582. Takumi Kawaguchi, Division of Gastroenterology, Department of Medicine,
Kurume University School of Medicine, Kurume, Japan.
Acknowledgments Vincent Wai-Sun Wong, Department of Medicine and Therapeutics, The
Members of the International Consensus Panel: Chinese University of Hong Kong, Hong Kong.
Arun Sanyal, Virginia Commonwealth University School of Medicine, Vlad Ratziu, Sorbonne Université, Assistance Publique-Hôpitaux de Paris,
Richmond, Virginia. Hôpital Pitié Salpêtrière, Institute of Cardiometabolism and Nutrition (ICAN),
Brent Neuschwander-Tetri, Division of Gastroenterology and Hepatology, Paris, France.
Saint Louis University, St. Louis, Missouri. Yusuf Yilmaz, Department of Gastroenterology, School of Medicine,
Claudio Tiribelli, Liver Center, Italian Liver Foundation, Trieste, Italy. Marmara University, Istanbul, Turkey; Institute of Gastroenterology, Marmara
David E. Kleiner, Laboratory of Pathology, Center for Cancer Research, University, Istanbul, Turkey.
National Cancer Institute, Bethesda, Maryland. Zobair Younossi, Center for Liver Diseases, Department of Medicine, Inova
Elizabeth Brunt, Department of Pathology and Immunology Washington Fairfax Hospital, Falls Church, Virginia.
University School of Medicine, St, Louis, Missouri.
Elisabetta Bugianesi, Division of Gastroenterology and Hepatology, Conflicts of interest
Department of Medical Sciences, University of Turin, Turin, Italy. The authors disclose the following: Rajiv Jalan, on the international consensus
Hannele Yki-Järvinen, Department of Medicine, University of Helsinki and panel, has research collaborations with Takeda and Yaqrit, and consults for
Helsinki University Hospital, and Minerva Foundation Institute for Medical Akaza and Yaqrit. Rajiv Jalan is the founder of Yaqrit Limited, which is
Research, Helsinki, Finland. developing UCL inventions for treatment of patients with cirrhosis. Rajiv
Henning Grønbæk, Department of Hepatology and Gastroenterology, Aarhus Jalan is an inventor of ornithine phenylacetate, which was licensed by UCL
University Hospital, Aarhus, Denmark. to Mallinckrodt. He is also the inventor of Yaq-001, DIALIVE, and Yaq-005,
Helena Cortez-Pinto, Clínica Universitária de Gastrenterologia, Laboratório the patents for which have been licensed by his university into a UCL
de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Portugal. spinout company, Yaqrit Ltd. The rest of authors declare no competing
Jacob George, Storr Liver Centre, Westmead Institute for Medical Research, interests for this manuscript. The remaining authors disclose no conflicts.
Westmead Hospital and University of Sydney, NSW, Australia
Jiangao Fan, Center for Fatty Liver, Department of Gastroenterology, Xin Funding
Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Mohammed Eslam and Jacob George are supported by the Robert W. Storr
Shanghai, China. Bequest to the Sydney Medical Foundation, University of Sydney; a National
Luca Valenti, Department of Pathophysiology and Transplantation, Health and Medical Research Council of Australia (NHMRC) Program Grant
Università degli Studi di Milano, and Translational Medicine, Department of (APP1053206, APP1149976); and Project Grants (APP1107178 and
Transfusion Medicine and Hematology, Fondazione IRCCS Ca’ Granda APP1108422). Philip N. Newsome, on the international consensus panel, is
Ospedale Maggiore Policlinico, Milano, Italy. funded and supported by the National Institute for Health Research (NIHR)
Manal Abdelmalek, Department of Medicine, Duke University, Durham, North Birmingham Biomedical Research Centre at the University Hospitals
Carolina. Birmingham NHS Foundation Trust and the University of Birmingham. The
Manuel Romero-Gomez, Hospital Universitario Virgen del Rocío de Sevilla, views expressed are those of the authors and not necessarily those of the
Instituto de Biomedicina de Sevilla, Biomedical Research Networking Center NIHR, the Department of Health and Social Care or the NHS.
in Hepatic and Digestive Diseases, Sevilla, Spain.
May 2020 MAFLD: A Proposed Nomenclature 2014.e1

Supplementary Figure 1. Conceptual framework of meta-


bolic dysfunction and pathogenesis of MAFLD. For metabolic
homeostasis, the neuroendocrine axes elicit multiple and
complex responses that orchestrate with caloric intake,
muscle mass, and physical activity, as well as with the
enterohepatic circulation, including gut microbiota, bile acids,
and their metabolites. These circles are interconnected at
various levels. For example, adiponectin signaling from adi-
pose tissue to liver, the liver (fibroblast growth factor 21) to
the central nervous system, the duodenum (cholecystokinin)
to the brain, and so on. These various inputs are integrated in
the liver. Dysfunctional homeostatic responses at any of
multiple levels are implicated in the heterogeneous patho-
genesis of MAFLD. CNS, central nervous system.

Supplementary Table 1.Results of Voting on Updating the Nomenclature of Fatty Liver Disease
1. Metabolic Associated Fatty Liver ± Disease (MAFL/MAFLD) (supported by 72.4% of participants)
2. Metabolic Fatty Liver ± Disease (MEFL/MEFLD) (supported by 17.2%)
3. Other suggestions (supported by 10.4%)

Supplementary Table 2.Research Gaps and Future Directions


Metabolic associated fatty liver disease (MAFLD) diagnosis and classification:
1) Define diagnostic criteria for MAFLD.
2) Define criteria for diagnosing MAFLD in the context of a second liver disease, for example, the level of alcohol consumption used to
distinguish MAFLD from dual MAFLD and alcohol-associated liver disease.
3) Should MAFLD be classified based on disease activity and stage rather than as a dichotomous state based on steatohepatitis and non-
steatohepatitis MAFLD.
Deconvolution of MAFLD heterogeneity
Research to define MAFLD subtypes.
Implications of the update on nomenclature
Implications of this update on clinical practice, the International Classification of Diseases systems and Disease Related Groups, and public
health policy.

You might also like