ArticlePDF AvailableLiterature Review

NAFLD as the metabolic hallmark of obesity

Authors:

Abstract

Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, associated with a high risk of progression to NASH, liver cirrhosis and hepatocarcinoma. Its prevalence is closely related to obesity (understood as adipose-based disease and insulin resistance), which makes that at present NAFLD can be considered a metabolic dysfunction hallmark, regardless of the body mass index. Despite being such a prevalent condition, with such severe consequences, at present there are no reliable biomarkers for its diagnosis or specific treatment. Significant and sustained weight loss, as well as some antidiabetic treatments, has shown promising results for NAFLD but data needs confirmation in larger clinical trials and longer follow-up. Efforts should be made for a better and more accurate baseline diagnosis (including large-scale genetics), identification of patients at higher risk for progression to NASH as well as adequate treatment, to allow us to offer a personalized approach in NAFLD in the context of precision medicine.
Vol.:(0123456789)
1 3
Internal and Emergency Medicine
https://doi.org/10.1007/s11739-022-03139-x
IM - REVIEW
NAFLD asthemetabolic hallmark ofobesity
AlbaRojano1· ElenaSena2· RamiroManzano‑Nuñez2· JuanM.Pericàs2,3· AndreeaCiudin1,4,5,6
Received: 16 August 2022 / Accepted: 17 October 2022
© The Author(s), under exclusive licence to Società Italiana di Medicina Interna (SIMI) 2022
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, associated with a
high risk of progression to NASH, liver cirrhosis and hepatocarcinoma. Its prevalence is closely related to obesity (understood
as adipose-based disease and insulin resistance), which makes that at present NAFLD can be considered a metabolic dysfunc-
tion hallmark, regardless of the body mass index. Despite being such a prevalent condition, with such severe consequences,
at present there are no reliable biomarkers for its diagnosis or specific treatment. Significant and sustained weight loss, as
well as some antidiabetic treatments, has shown promising results for NAFLD but data needs confirmation in larger clinical
trials and longer follow-up. Efforts should be made for a better and more accurate baseline diagnosis (including large-scale
genetics), identification of patients at higher risk for progression to NASH as well as adequate treatment, to allow us to offer
a personalized approach in NAFLD in the context of precision medicine.
Keywords Obesity· Non-alcoholic fatty liver disease· Liver steatosis· Liver inflammation
Introduction
Non-alcoholic fatty liver disease (NAFLD) has become the
most common chronic liver disease worldwide and affects
25–35% of adults in the general population from western
countries [1].
Additionally, the incidences of obesity and metabolic
syndrome have exponentially increased in recent years,
reaching epidemic proportions [2]. The latest report of the
World Health Organization in 2016 estimated that 650 mil-
lion adults were living with obesity, meaning a prevalence
of 13% of the overall adult population [3].
NAFLD includes a spectrum of histopathological and
clinical features with a direct relationship with obesity,
insulin resistance (IR)/type 2 diabetes (T2D) and metabolic
syndrome (Met S) [4].
A pooled analysis from 200 countries published in 2006
showed that between 1980 and 2015 obesity prevalence dou-
bled in 73 countries [5] and this increase was linked to an
expansion from 37.4% in 2005 to 2010 to 41, 5% in 2016 to
2019 in NAFLD diagnoses in this adult subpopulation [6].
Furthermore, more recent data showed that when either obe-
sity or T2D are present the prevalence of NAFLD is about
60–80% [7] and when both conditions are present NAFLD
reaches 80–100% [8].
Alba Rojano and Elena Sena contributed equally to the manuscript.
* Juan M. Pericàs
pericasjm@gmail.com
* Andreea Ciudin
andreea.ciudin@vallhebron.cat
1 Endocrinology andNutrition Department, Vall
d’HebronInstitut de Recerca (VHIR), Hospital Universitari
Vall d’Hebron, Vall d’Hebron Barcelona Campus
Hospitalari, Barcelona, Spain
2 Liver Unit, Internal Medicine Department, Vall
d’HebronInstitut de Recerca (VHIR), Hospital Universitari
Vall d’Hebron, Vall d’Hebron Barcelona Campus
Hospitalari, Barcelona, Spain
3 Centro de Investigación Biomédica en Red de Enfermedades
Digestivas Y Hepáticas (CIBERehd), Instituto de Salud
Carlos III, Madrid, Spain
4 CIBER de Diabetes Y Enfermedades Metabólicas Asociadas
(CIBERDem), Instituto de Salud Carlos III, Madrid, Spain
5 Department ofCellular Biology, Physiology
andImmunology, Universitat Autònoma de Barcelona,
Barcelona, Spain
6 Pathology Department, Hospital Universitari Vall d’Hebron,
Vall d’Hebron Barcelona Campus Hospitalari, Barcelona,
Spain
Internal and Emergency Medicine
1 3
Moreover, at present, NAFLD is the most prevalent form
of chronic liver disease in childhood and adolescence, affect-
ing approximately 10%–20% of the general paediatric popu-
lation. NAFLD is expected to become the main cause of
liver failure and indication for liver transplantation in this
population in the Western world within the next 10years [9].
Similar to findings in adults, when overweight or obesity
was present, the prevalence of NAFLD was 6 to 26 times
higher, respectively, in children and adolescents between 6
and 18years of age [10].
Additionally, the leading cause of death among patients
with NAFLD remains cardiovascular disease, followed by
extra-hepatic cancers and liver-related complications [11,
12]. Furthermore, although it has not been widely demon-
strated, recent data have shown that NAFLD could be con-
sidered as a marker of subclinical atherosclerosis as well as
a strong cardiovascular risk factor even at a very early age
[13], reflecting the strong relationship between NAFLD and
MetS.
Insulin resistance (IR), one of the major components of
obesity and MetS [14] plays a key role in the pathophysi-
ology of NAFLD, by promoting the progression of simple
steatosis to liver inflammation and fibrosis, also known as
non-alcoholic steato-hepatitis (NASH) [15].
It is estimated that 20–30% of patients with NAFLD will
progress to non-alcoholic steatohepatitis (NASH), which is
characterized by hepatic inflammation and cellular injury,
with variable degrees of fibrosis [16]. Even though NAFLD
is usually asymptomatic in the early stages, the condition
can silently progress to advanced stages including cirrho-
sis, hepatocellular carcinoma (HCC) or liver failure [17].
Indeed, for patients with NASH-related cirrhosis has been
observed a > tenfold increase in risk for HCC [18]. Further-
more, NASH-related cirrhosis has become a leading cause
of liver transplantation in developed countries [19] Obesity
is one of the major risk factors for progression from NAFLD
to NASH [20]—Fig.1.
Additionally, in the last decade, several genetic risk fac-
tors have been identified and associated with the suscepti-
bility of NAFLD and progression to NASH and advanced
fibrosis [21]. Among them, single nucleotide polymorphism
(SNP) rs738409 of the patatin-like phospholipase domain-
containing protein 3 (PNPLA3) gene [22, 23] and SNP
rs58542926 of the transmembrane protein involved in mol-
ecule transport (TM6SF2) gene [24] have been identified in
several genome-wide association studies (GWAS) as risk
factors for progressive NASH and advanced liver fibrosis
[17].
Our group has recently shown that the presence of IR
on top of these polymorphisms significantly increased the
risk of NASH and severe fibrosis in patients with obesity
[25], highlighting the strong relationship between obesity
and MetS and NAFLD.
Due to the strong bidirectional association between MetS
and NAFLD, in 2020, worldwide expert consensus proposed
a more realistic concept of the disease, including a change
of the nomenclature of NAFLD to Metabolic dysfunction-
Associated Fatty Liver Disease (MAFLD), which now
requires, in addition to hepatic steatosis, the co-occurrence
of one of three features: 1) overweight or obesity, 2) T2DM,
or 3) lean or normal weight with evidence of metabolic dys-
regulation (2628). The general conception is that NAFLD
is no longer an isolated disease; but rather a condition that
appears in conjunction with other metabolic factors, being
considered, at present, the hepatic manifestation of MetS
[26, 27].
It should be noted that the criteria used in this consen-
sus to define “overweight”, “obesity” and “non-obese” sub-
jects is BMI. Classically, obesity is defined worldwide as
BMI > 30kg/m2 [29] and one of the criteria for MetS is
waist circumference [30] Furthermore, in several studies
so far BMI and waist circumference positively correlates
with NAFLD presence and progression [15]. Nevertheless,
from a mechanistically point of view, obesity is defined
as the excess of body fat, regardless of the BMI [31]. The
presence of NAFLD and MAFLD features in lean subjects
(BMI < 25kg/m2), brings to front a more recent debate,
regarding the accuracy of BMI in defining obesity and the
risk of its metabolic complications. The EASO position
statement in 2019 [32] proposes the new definition of obe-
sity as adiposity-based-chronic disease and Snitker etal.,
2010 defines obesity as %body fat > 25 in males and > 35%
in females [33].
NAFLD/MAFLD is the hallmark of the body fat content,
in terms of total amount, distribution and functionality [34].
Several studies associate NAFLD with body fat accu-
mulation and loss of muscle mass. Shi etal. showed that
visceral adipose tissue (VAT) was an independent predictor
of NAFLD regardless of obesity defined by BMI [35]. Fur-
thermore, VAT accumulation induces insulin resistance and
exacerbates liver damage in NAFLD [36]. Some studies also
showed that the presence of sarcopenia (muscle mass loss)
is associated with metabolic syndrome, and cardiovascular
disease and is a risk factor for non-alcoholic steatohepatitis
(NASH) and fibrosis (≥ F29 [37]. Nevertheless, recent data
reflected that the pathophysiology of NAFLD may be more
dependent on fat accumulation than on the loss of muscle
mass [38].
NAFLD histopathology inobesity
As explained before, NAFLD physiopathology is complex
and includes metabolic, environmental, genetic, and gut
microbial factors [39]. Insulin resistance, a hallmark of obe-
sity and excess body fat, plays a key role in the development
Internal and Emergency Medicine
1 3
of the histologic features found in NAFLD- Fig.2. IR
implies an impairment in the suppression of hepatic glucose
production that correlates with increased gluconeogenesis
and free fatty acid (FFA) levels and a decrease in glycogen
synthesis. These alterations in the metabolic cascades result
in lipid accumulation, which in turn results in a preferential
shift from carbohydrate to FFA beta-oxidation (15, 4042).
Additionally, experimental studies have shown a reduced
Fig. 1 NAFLD the metabolic hallmark in obesity. FFAs: free fatty acids, NAFLD: non-alcoholic fatty liver disease
Internal and Emergency Medicine
1 3
synthesis and secretion of very low-density lipoprotein
(VLDL) and excessive importation of FFA from adipose
tissue [43]. This lipid accumulation has been linked as a
trigger in the activation of several inflammatory pathways
related to pro-inflammatory cytokines, such as TNFα or
IL¬6 [44]. All these physiopathological changes will finally
end in an increase in oxidative stress and mitochondrial leak-
age, resulting in hepatic fibrosis and promote more insulin
resistance [45]. Recent data in fatty liver models without
fibrosis have shown that lipid accumulation associated with
obesity induces an architectural distortion, resulting in: a)
reduced sinusoidal space, b) increased intrahepatic vascular
resistance and c) potentially portal hypertension related to
obesity (PH) which will contribute to the progression to liver
cirrhosis and hemodynamic decompensation (4648).
NAFLD diagnosis inobesity
The dramatic increase in the prevalence and impact of
NAFLD has led to the rapid development of new diagnostic
methods.
As explained before, the presence of liver steatosis (diag-
nosed by imaging, biochemical scores or liver biopsy) and
obesity will automatically convert into MAFLD diagnosis,
according to the latest consensus [26]. Nevertheless, at present
there is no effective screening method with high sensibility
and low false positives to be considered the gold-standard.
In this regard, guidelines such as the American Association
for the Study of Liver Diseases (AASLD) does not recom-
mend population screening for NAFLD [49]. By contrast,
EASL–EASD–EASO Clinical Practice Guidelines in 2016
recommend non-invasive screening to predict steatosis, NASH
and fibrosis in all the subjects living with obesity and elevated
liver enzymes. If ultrasound steatosis or high-risk fibrosis
markers are obtained a referral to hepatologist is required [50].
More recent recommendation, propose a step-by-step
approach: (5052) to diagnose NAFLD based on: 1) the use
of serologic non-invasive tests with high negative predictive
value to exclude the presence of significant liver fibrosis, 2)
the performance of a transient elastography to identify patients
with suspicion of liver fibrosis and 3) the indication of liver
biopsy to accurately stratify the disease and for consideration
in pharmacological clinical trials.
Fig. 2 Physiopathological mechanisms that link obesity with NAFLD
Internal and Emergency Medicine
1 3
Non‑invasive biomarkers
Diagnosis ofhepatic steatosis ‑NAFLD
Serum biomarkers
Increased levels of ALT, AST, or γ-glutamyltransferase
(GGT) are used to diagnose NAFLD. Nevertheless,
although AST levels were associated with NASH in
advanced fibrosis, most patients with NASH or advanced
fibrosis had normal AST levels [53] and these values may
differ with age, sex, and ethnical origin [54].
Liver imaging
Abdominal ultrasound is usually the first diagnostic tool per-
formed to assess hepatic steatosis. Ultrasound devices are
widely available in primary and specialized care, it is easy
to perform and provides additional information (i.e., liver
structure, focal lesions or indirect signs of portal hyperten-
sion). However, hepatic ultrasound shows a low sensitivity
to detect mild steatosis, especially in subjects with obesity
and it is operator-dependent. In this regard, several serologic
non-invasive test has been developed in the recent years,
such as Hepatic Steatosis Index (HIS) and Fatty Liver Index
(FLI), which include usual blood test parameters and meta-
bolic data to detect fatty liver disease [55, 56].
The Controlled Attenuation Parameter (CAP) software
has been recently incorporated into transient elastography
devices. CAP allows a quantitative measurement of liver
steatosis with higher diagnostic performance than liver
ultrasound and serologic tests. [57].
Diagnosis ofliver fibrosis
Serum biomarkers
The most validated serologic fibrosis score for NAFLD are
FIB-4 index and NAFLD Fibrosis Score (NFS). Both test
employ usual demographic and analytical parameters (i.e.,
age and liver enzymes) and had a high negative predictive
value (> 90%) to exclude advanced fibrosis with the low-
est threshold [58]. Developed by Siemens, the Enhanced
Liver Fibrosis (ELFTM) panel predicts advanced fibro-
sis by measuring hyaluronic acid (HA), procollagen III
amino-terminal peptide (PIIINP), and the tissue inhibi-
tor of matrix metalloproteinase 1 (TIMP-1) and has been
implemented in some European guidelines [59].
Liver imaging
Transient elastography (TE) (Fibroscan®) is a ultrasound-
based technique for diagnosing liver fibrosis with adequate
diagnostic accuracy [60, 61]. Besides, TE values (liver stiff-
ness measurement-LSM- and controlled attenuation param-
eter) have been associated with the risk of developing clini-
cally significant portal hypertension (CSPH) and clinical
events [62, 63]. However, TE accuracy for liver fibrosis and
CSPH estimation significantly decreases in patients with obe-
sity, which has been proved independently of the liver disease
etiology but it is more pronounced in NASH patients. Two-
dimensional shear wave elastography (2D-SWE), shear wave
dispersion slope (SWDS), and Attenuation Imaging (ATI) are
novel techniques that quantify liver stiffness, steatohepatitis,
and steatosis of hepatic tissue, respectively, and are installed
in modern ultrasound devices. Nonetheless, only a few stud-
ies have assessed the accuracy of 2D-SWE, SWDS, and ATI
in NAFLD patients, and cutoff values are still not validated
[64]. These techniques have not been evaluated extensively
for NASH diagnosis, and, therefore, TE continues to be the
primary imaging technique to assess liver stiffness.
Magnetic resonance imaging (MRI) can measure hepatic
steatosis and fibrosis through the proton density fat frac-
tion (PDFF) software and magnetic resonance elastography
(MRE), respectively. These methods have been proven the
most accurate methods for steatosis and fibrosis detection
and staging. Nonetheless, the high cost and restricted avail-
ability (including size in patients with obesity) limit their
implementation in clinical practice and are mainly used in
clinical trials and research studies.
Liver biopsy
Liver Biopsy remains as the gold standard technique for the
diagnosis of NAFLD and for disease staging and progres-
sion of NASH (to assess ballooning and inflammation) NAS
score and CRN criteria are the most extended histological
panels to diagnose NAFLD [65]. However, due to the inva-
sive nature of the procedure, the indication is limited to
selected cases. Furthermore, in patients with obesity, the
technical difficulty, and the ability to obtain high-quality
samples might be hampered according to the width of sub-
cutaneous fat.
NAFLD management andtreatment
inobesity
Despite being a prevalent condition and a public health
concern, currently there are no pharmacological treatments
approved for NAFLD/MAFLD. New pharmacotherapy
that targets metabolic hepatic pathways, such as bile acid
Internal and Emergency Medicine
1 3
metabolism, mitochondrial lipogenesis and gluconeogenesis
and fatty acid synthesis that have demonstrated the preven-
tion of apoptosis, inflammation, and fibrosis are being stud-
ied (6669)
The current intervention in NAFLD patients is mainly
centered in weight loss and in improving metabolic condi-
tion, such as IR and/or T2D [70, 71]. It has been shown
that a 7–10% of total weight loss can improve histological
NAFLD features and progression to NASH [72]. Addition-
ally, some antidiabetic drugs have demonstrated resolution
of the histological lesions defining NASH with no worsen-
ing of fibrosis (7375). Nevertheless, if there is a strong
suspicion of a more advanced liver disease, patients should
be referred to a liver specialist (hepatologist or gastroenter-
ologist), as stated by current guidelines, based on blood and
imaging tests. [70]—Table1.
Dietary andlifestyle changes
Several studies examined the role of weight loss on NAFLD
progression, confirming the tight relationship between obe-
sity and liver affectation [76]. A total loss of 5–7% percent
of body weight demonstrated an improvement in hepatic
fibrosis and steatosis [77, 78]. Studies based on a calorie-
restriction diet demonstrated a decrease in intrahepatic fat
content and inflammation corresponding to insulin sensi-
tivity improvement [77, 78]. Furthermore, not only calorie
restriction but changes in diet composition, such as the Med-
iterranean diet have demonstrated improvement in NAFLD
outcomes and IR [79].
Pharmacological therapy
At present, there are no FDA or EMA-approved drugs for
NAFLD. We will refer to those drugs used for T2D and/or
obesity that significantly impact on NAFLD, such as Gluca-
gon-like peptide-1 receptor agonist (GLP-1 RA), thiazolidin-
edione and SGLT2 inhibitors.
GLP-1 RAs improve glycaemic control, facilitate
remarkable and maintained body weight loss as well as car-
diovascular beneficial effects (8082). Furthermore, in these
studies a beneficial effect on NAFLD was seen, suggesting
once more that NAFLD is the liver complication of obe-
sity and metabolic syndrome. In the LEAN study, [83] 39%
patients with T2D and obesity achieved NASH resolution
with subcutaneous 1.8mg daily Liraglutide. Additionally,
the results of a phase-2 trial including 320 patients with
obesity and biopsy-proven NASH revealed that semaglu-
tide was superior to placebo in terms of NASH resolution.
[84]. The recent guideline from the American Association
of Clinical Endocrinologists (AACE) recommends treatment
with liraglutide 3mg daily for the management of NAFLD
in patients with obesity. [70].
Thiazolidinediones. Pioglitazone and rosiglitazone
improve peripheral insulin sensitivity and are potent acti-
vators of the nuclear receptor PPARγ implied in adipocyte
differentiation and lipid and glucose metabolism, decreasing
hepatic lip content [85, 86]. Furthermore, thiazolidinediones
have demonstrated a reduction of hepatic stellate cell activa-
tion and fibrosis experimental models [87]. Pioglitazone has
demonstrated also reductions in inflammation and hepato-
cyte degeneration as well as in fibrosis [88].
Inhibitors of SGLT2 (canagliflozin, dapagliflozin, empa-
gliflozin and ertugliflozin) lead to the increased urinary
excretion of glucose. iSGLT2 have demonstrated in numer-
ous trials the reduction of the risk of major cardiovascular
events [8991]. Also have demonstrated in animal models
improvement in levels of liver enzymes, steatosis, hepato-
cyte damage and fibrosis [92] but there is a lack of clinical
efficacy in RCT [74, 93].
At present, there are several experimental studies with
other pharmacotherapy groups that target hepatic alterations
of metabolism in NAFLD that have demonstrated efficacy,
but a small number were tested in clinical trials. Some of the
new clinical phase III trials include inhibitors of ketohexoki-
nase that block the final step of fructose metabolism inhibit-
ing lipogenesis [67]. Mitochondrial pyruvate carrier (MPC)
inhibitors prevent the import of pyruvate into the mitochon-
dria, blocking gluconeogenesis and fatty acid synthesis path-
ways [68, 69]. Lastly, inhibitors of enzymes involved in the
synthesis of triglycerides s such as acetyl-CoA carboxylase
a and Stearoyl-CoA desaturase have also demonstrated pre-
vention in apoptosis, inflammation, and fibrosis [94, 95].
Metabolic procedural techniques: bariatric
endoscopy andbariatric surgery
Bariatric surgery and bariatric endoscopy are valid treat-
ment options for patients with obesity, especially those
that associate MeTs. At present, there is limited evidence
Table 1 Criteria for referral to liver specialist (Hepatologist or Gas-
troenterologist)
FIB-4 index for liver fibrosis, LSM liver stiffness measurement, ELF
enhanced liver fibrosis
Parameter Intermediate risk High risk
Blood transaminases level Elevated Elevated
Liver steatosis on imaging Present Present
FIB-4 1.3–2.67 2.67
LSM (kPa) 8–12 > 12
ELF 7.7–9.8 > 9.8
Internal and Emergency Medicine
1 3
for the use of endoscopic metabolic techniques in patients
with NAFLD and obesity. Some studies have shown his-
tological improvements in small series [96]. Bariatric
endoscopy is not currently incorporated in clinical prac-
tice guidelines but could be considered in highly selected
patients unable to undergo bariatric surgery in experienced
centers.
Bariatric surgery (BS) has been proven to induce signifi-
cant weight loss and potentially positively impact NAFLD
and NASH outcomes, improving necrosis, inflammation
and fibrosis in a longitudinal prospective study with serial
liver biopsies, (9799). Nevertheless, the methodology of
these studies is heterogeneous and at present there is no
randomized clinical trial aimed to shed light on this issue.
Furthermore, recent data suggested worsening of NAFLD/
NASH features in some patients following BS [100]. On
these bases, we consider that the indication for BS in patients
with NAFLD/NASH should be individualized and carefully
analyzed in each patient.
Concluding remarks
Non-alcoholic fatty liver disease (NAFLD) has become the
most common chronic liver disease worldwide, associated
with a high risk of progression to NASH, liver cirrhosis and
hepatocarcinoma. Its prevalence is closely related to obesity
(understood as adipose-based disease and insulin resistance),
which makes that at present NAFLD can be considered a
metabolic dysfunction hallmark, regardless of the body mass
index. Despite being such a prevalent condition, with such
severe consequences, at present there are no reliable bio-
markers for its diagnosis or specific treatment. Significant
and sustained weight loss, as well as some antidiabetic treat-
ments, has shown promising results for NAFLD. Neverthe-
less, the actual data needs confirmation in larger clinical
trials and longer follow-up. Efforts should be made for a
better and accurate baseline diagnosis (including large-scale
genetics), identification of patients at higher risk for progres-
sion to NASH as well as adequate treatment, to allow us to
offer a personalized approach in NAFLD in the context of
precision medicine.
Declarations
Conflict of interest Authors declare no conflict of interest related to
this work.
Human and animal rights statement This article does not contain any
studies with human participants or animals performed by any of the
authors.
Informed consent For this type of study formal consent is not required.
References
1. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L,
Wymer M (2016) Global epidemiology of nonalcoholic fatty
liver disease-meta-analytic assessment of prevalence. Incidence
Outcomes Hepatol 64(1):73–84. https:// doi. org/ 10. 1002/ hep.
28431
2. The GBD 2015 Obesity Collaborators (2015) Obesity Collabo-
rators. (2017) Health Effects of Overweight and Obesity in 195
Countries over 25 Years. N Engl J Med 377:13–27. https:// doi.
org/ 10. 1056/ NEJMo a1614 362
3. Obesity and overweight. https:// www. who. int/ news- room/ fact-
sheets/ detail/ obesi ty- and- overw eight (accessed 2022–08–11).
4. Buzzetti E, Pinzani M, Tsochatzis EA (2016) The multiple-hit
pathogenesis of non-alcoholic fatty liver disease (NAFLD).
Metabolism 65(8):1038–1048. https:// doi. org/ 10. 1016/j. metab
ol. 2015. 12. 012
5. NCD Risk Factor Collaboration (NCD-RisC) (2016) Trends in
adult body-mass index in 200 countries from 1975 to 2014: a
pooled analysis of 1698 population-based measurement studies
with 19·2 million participants. Lancet 387:1377–1396. https://
doi. org/ 10. 1016/ S0140- 6736(16) 30054-X
6. Cholongitas E, Pavlopoulou I, Papatheodoridi M, Markakis GE,
Bouras E, Haidich A-B, Papatheodoridis G (2021) Epidemiology
of nonalcoholic fatty liver disease in europe: a systematic review
and meta-analysis. Ann Gastroenterol 34:404–414
7. Loomba R, Abraham M, Unalp A, Wilson L, Lavine J, Doo
E, Bass NM (2012) The nonalcoholic steatohepatitis clinical
research network. Association between diabetes, family history
of diabetes, and risk of nonalcoholic steatohepatitis and fibrosis.
Hepatology 56:943–951. https:// doi. org/ 10. 1002/ hep. 25772
8. Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contre-
ras M, Landt CL, Harrison SA (2011) Prevalence of nonalco-
holic fatty liver disease and nonalcoholic steatohepatitis among
a largely middle-aged population utilizing ultrasound and liver
biopsy: a prospective study. Gastroenterology 140(1):124–131.
https:// doi. org/ 10. 1053/j. gastro. 2010. 09. 038
9. Berardis S, Sokal E (2014) Pediatric non-alcoholic fatty
liver disease: an increasing public health issue. Eur J Pediatr
173(2):131–139
10. Sharma V, Coleman S, Nixon J, Sharples L, Hamilton-Shield
J, Rutter H, Bryant M (2019) A systematic review and meta-
analysis estimating the population prevalence of comorbidi-
ties in children and adolescents aged 5 to 18 years. Obes Rev
20(10):1341–1349. https:// doi. org/ 10. 1111/ obr. 12904
11. Byrne CD, Targher G (2015) NAFLD: a multisystem disease. J
Hepatol 62(1 Suppl):S47-64. https:// doi. org/ 10. 1016/j. jhep. 2014.
12. 012
12. Targher G, Tilg H, Byrne CD (2021) Non-alcoholic fatty liver
disease: a multisystem disease requiring a multidisciplinary and
holistic approach. Lancet Gastroenterol Hepatol 6(7):578–588.
https:// doi. org/ 10. 1016/ S2468- 1253(21) 00020-0
13. Di Sessa A, Umano GR, Miraglia Del Giudice E (2017) The
association between non-alcoholic fatty liver disease and cardio-
vascular risk in children. Children (Basel) 4(7):E57. https:// doi.
org/ 10. 3390/ child ren40 70057
14. Roberts CK, Hevener AL, James Barnard R (2013) Metabolic
syndrome and insulin resistance: underlying causes and modifi-
cation by exercise training. Compr Physiol 3:1–58. https:// doi.
org/ 10. 1002/ cphy. c1100 62
15. Marchesini G, Brizi M, Bianchi G, Tomassetti S, Bugianesi E,
Lenzi M, McCullough AJ, Natale S, Forlani G, Melchionda N
(2001) Nonalcoholic fatty liver disease: a feature of the metabolic
syndrome. Diabetes 50(8):1844–1850. https:// doi. org/ 10. 2337/
diabe tes. 50.8. 1844
Internal and Emergency Medicine
1 3
16. Liu W, Baker RD, Bhatia T, Zhu L, Baker SS (2016) Patho-
genesis of nonalcoholic steatohepatitis. Cell Mol Life Sci
73(10):1969–1987. https:// doi. org/ 10. 1007/ s00018- 016- 2161-x
17. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ
(2018) Mechanisms of NAFLD development and therapeutic
strategies. Nat Med 24(7):908–922. https:// doi. org/ 10. 1038/
s41591- 018- 0104-9
18. Dyson J, Jaques B, Chattopadyhay D, Lochan R, Graham J, Das
D, Aslam T, Patanwala I, Gaggar S, Cole M, Sumpter K, Stewart
S, Rose J, Hudson M, Manas D, Reeves HL (2014) Hepatocel-
lular cancer: the impact of obesity, type 2 diabetes and a multi-
disciplinary team. J Hepatol 60(1):110–117. https:// doi. org/ 10.
1016/j. jhep. 2013. 08. 011
19. Pais R, Barritt AS, Calmus Y, Scatton O, Runge T, Lebray P,
Poynard T, Ratziu V, Conti F (2016) NAFLD and liver trans-
plantation: current burden and expected challenges. J Hepatol
65(6):1245–1257. https:// doi. org/ 10. 1016/j. jhep. 2016. 07. 033
20. Kim Y, Chang Y, Cho YK, Ahn J, Shin H, Ryu S (2019) Obesity
and weight gain are associated with progression of fibrosis in
patients with nonalcoholic fatty liver disease. Clin Gastroenterol
Hepatol 17:543–550. https:// doi. org/ 10. 1016/j. cgh. 2018. 07. 006
21. Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam
M, George J, Bugianesi E (2018) Global burden of NAFLD and
NASH: trends, predictions, risk factors and prevention. Nat Rev
Gastroenterol Hepatol 15(1):11–20. https:// doi. org/ 10. 1038/ nrgas
tro. 2017. 109
22. Míková I, Neroldová M, Hubácek JA, Dlouhá D, Jirsa M, Hons-
ová E, Sticová E, Lánská V, Špicák J, Trunecka P (2020) Donor
PNPLA3 and TM6SF2 variant alleles confer additive risks
for graft steatosis after liver transplantation. Transplantation
104(3):526–534. https:// doi. org/ 10. 1097/ TP. 00000 00000 002876
23. Rinaldi L, Pafundi PC, Galiero R, Caturano A, Morone MV, Sil-
vestri C, Giordano M, Salvatore T, Sasso FC (2021) Mechanisms
of non-alcoholic fatty liver disease in the metabolic syndrome.
A Narrative Review Antioxidants 10(2):270. https:// doi. org/ 10.
3390/ antio x1002 0270
24. Valenti L, Al-Serri A, Daly AK, Galmozzi E, Rametta R, Don-
giovanni P, Nobili V, Mozzi E, Roviaro G, Vanni E, Bugianesi
E, Maggioni M, Fracanzani AL, Fargion S, Day CP (2010)
Homozygosity for the patatin-like phospholipase-3/adiponutrin
i148m polymorphism influences liver fibrosis in patients with
nonalcoholic fatty liver disease. Hepatology 51(4):1209–1217.
https:// doi. org/ 10. 1002/ hep. 23622
25. Gabriel-Medina P, Ferrer-Costa R, Rodriguez-Frias F, Ciudin
A, Augustin S, Rivera-Esteban J, Pericàs JM, Selva DM (2022)
Influence of type 2 diabetes in the association of PNPLA3
Rs738409 and TM6SF2 Rs58542926 polymorphisms in NASH
advanced liver fibrosis. Biomedicines 10(5):1015. https:// doi. org/
10. 3390/ biome dicin es100 51015
26. Eslam M, Sanyal AJ, George J (2020) International consen-
sus panel. mafld: a consensus-driven proposed nomenclature
for metabolic associated fatty liver disease. Gastroenterology
158:1999–2014. https:// doi. org/ 10. 1053/j. gastro. 2019. 11. 312
27. Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G,
Romero-Gomez M, Zelber-Sagi S, Wong VW-S, Dufour J-F,
Schattenberg JM, Kawaguchi T, Arrese M, Valenti L, Shiha G,
Tiribelli C, Yki-Järvinen H, Fan J-G, Grønbæk H, Yilmaz Y,
Cortez-Pinto H, Oliveira CP, Bedossa P, Adams LA, Zheng M-H,
Fouad Y, Chan W-K, Mendez-Sanchez N, Ahn SH, Castera L,
Bugianesi E, Ratziu V, George J (2020) A New definition for
metabolic dysfunction-associated fatty liver disease: an inter-
national expert consensus statement. J Hepatol 73(1):202–209.
https:// doi. org/ 10. 1016/j. jhep. 2020. 03. 039
28. Tilg H, Effenberger M (2020) From NAFLD to MAFLD: when
pathophysiology succeeds. Nat Rev Gastroenterol Hepatol
17(7):387–388. https:// doi. org/ 10. 1038/ s41575- 020- 0316-6
29. Apovian CM (2016) Obesity: definition, comorbidities, causes,
and burden. Am J Manag Care 22(7 Suppl):s176-185
30. Alberti KG, Zimmet PZ (1998) Definition, diagnosis and clas-
sification of diabetes mellitus and Its complications. part 1:
diagnosis and classification of diabetes mellitus provisional
report of a who consultation. Diabet Med 15:539–553
31. Mechanick JI, Hurley DL, Garvey WT (2017) Adiposity-based
chronic disease as a new diagnostic term: The American Asso-
ciation of Clinical Endocrinologists and American College of
Endocrinology Position Statement. Endocr Pract 23(3):372–
378. https:// doi. org/ 10. 4158/ EP161 688. PS
32. Frühbeck G, Busetto L, Dicker D, Yumuk V, Goossens GH,
Hebebrand J, Halford JGC, Farpour-Lambert NJ, Blaak EE,
Woodward E, Toplak H (2019) The ABCD of obesity: an
EASO position statement on a diagnostic term with clinical
and scientific implications. Obes Facts 12(2):131–136. https://
doi. org/ 10. 1159/ 00049 7124
33. Snitker S (2010) Use of body fatness cutoff points. Mayo Clin
Proc 85:1057–1058. https:// doi. org/ 10. 4065/ mcp. 2010. 0583
34. Garvey WT (2022) Is obesity or adiposity-based chronic
disease curable: the set point theory, the environment, and
second-generation medications. Endocr Pract 28(2):214–222.
https:// doi. org/ 10. 1016/j. eprac. 2021. 11. 082
35. Shi Y-X, Chen X-Y, Qiu H-N, Jiang W-R, Zhang M-Y, Huang
Y-P, Ji Y-P, Zhang S, Li C-J, Lin J-N (2021) Visceral fat area to
appendicular muscle mass ratio as a predictor for nonalcoholic
fatty liver disease independent of obesity. Scand J Gastroen-
terol 56(3):312–320. https:// doi. org/ 10. 1080/ 00365 521. 2021.
18792 44
36. Malaguarnera M, Di Rosa M, Nicoletti F, Malaguarnera L
(2009) Molecular mechanisms involved in NAFLD progres-
sion. J Mol Med (Berl) 87(7):679–695. https:// doi. org/ 10. 1007/
s00109- 009- 0464-1
37. Kim JA, Choi KM (2019) Sarcopenia and fatty liver dis-
ease. Hepatol Int 13(6):674–687. https:// doi. org/ 10. 1007/
s12072- 019- 09996-7
38. Miyake T, Miyazaki M, Yoshida O, Kanzaki S, Nakaguchi
H, Nakamura Y, Watanabe T, Yamamoto Y, Koizumi Y,
Tokumoto Y, Hirooka M, Furukawa S, Takeshita E, Kum-
agi T, Ikeda Y, Abe M, Toshimitsu K, Matsuura B, Hiasa Y
(2021) Relationship between body composition and the his-
tology of non-alcoholic fatty liver disease: a cross-sectional
study. BMC Gastroenterol 21(1):170. https:// doi. org/ 10. 1186/
s12876- 021- 01748-y
39. Eslam M, Valenti L, Romeo S (2018) Genetics and epigenetics of
NAFLD and NASH: clinical impact. J Hepatol 68(2):268–279.
https:// doi. org/ 10. 1016/j. jhep. 2017. 09. 003
40. Krssak M, Brehm A, Bernroider E, Anderwald C, Nowotny P,
Dalla Man C, Cobelli C, Cline GW, Shulman GI, Waldhäusl
W, Roden M (2004) Alterations in postprandial hepatic glyco-
gen metabolism in type 2 diabetes. Diabetes 53(12):3048–3056.
https:// doi. org/ 10. 2337/ diabe tes. 53. 12. 3048
41. Lomonaco R, Bril F, Portillo-Sanchez P, Ortiz-Lopez C, Orsak B,
Biernacki D, Lo M, Suman A, Weber MH, Cusi K (2016) Meta-
bolic impact of nonalcoholic steatohepatitis in obese patients
with type 2 diabetes. Diabetes Care 39(4):632–638. https:// doi.
org/ 10. 2337/ dc15- 1876
42. Utzschneider KM, Van de Lagemaat A, Faulenbach MV, Goe-
decke JH, Carr DB, Boyko EJ, Fujimoto WY, Kahn SE (2010)
Insulin resistance is the best predictor of the metabolic syndrome
in subjects with a first-degree relative with type 2 diabetes. Obe-
sity (Silver Spring) 18(9):1781–1787. https:// doi. org/ 10. 1038/
oby. 2010. 77
43. Heeren J, Scheja L (2021) Metabolic-associated fatty liver dis-
ease and lipoprotein metabolism. Mol Metab 50:101238. https://
doi. org/ 10. 1016/j. molmet. 2021. 101238
Internal and Emergency Medicine
1 3
44. Tilg H, Diehl AM (2000) Cytokines in alcoholic and nonal-
coholic steatohepatitis. N Engl J Med 343(20):1467–1476.
https:// doi. org/ 10. 1056/ NEJM2 00011 16343 2007
45. Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, Con-
tos MJ, Sterling RK, Luketic VA, Shiffman ML, Clore JN
(2001) Nonalcoholic steatohepatitis: association of insulin
resistance and mitochondrial abnormalities. Gastroenterology
120(5):1183–1192. https:// doi. org/ 10. 1053/ gast. 2001. 23256
46. Ryou M, Stylopoulos N, Baffy G (2020) Nonalcoholic fatty
liver disease and portal hypertension. Explor Med 1:149–169
47. Francque S, Verrijken A, Mertens I, Hubens G, Van Marck E,
Pelckmans P, Van Gaal L, Michielsen P (2010) Noncirrhotic
human nonalcoholic fatty liver disease induces portal hyper-
tension in relation to the histological degree of steatosis. Eur
J Gastroenterol Hepatol 22(12):1449–1457. https:// doi. org/ 10.
1097/ MEG. 0b013 e3283 3f14a1
48. Pasarín M, La Mura V, Gracia-Sancho J, García-Calderó H,
Rodríguez-Vilarrupla A, García-Pagán JC, Bosch J, Abraldes
JG (2012) Sinusoidal endothelial dysfunction precedes
inflammation and fibrosis in a model of NAFLD. PLoS ONE
7(4):e32785. https:// doi. org/ 10. 1371/ journ al. pone. 00327 85
49. Corey KE, Klebanoff MJ, Tramontano AC, Chung RT, Hur
C (2016) Screening for Nonalcoholic steatohepatitis in
individuals with type 2 diabetes: a cost-effectiveness analy-
sis. Dig Dis Sci 61(7):2108–2117. https:// doi. org/ 10. 1007/
s10620- 016- 4044-2
50. (2016) EASL–EASD–EASO Clinical Practice Guidelines for the
Management of Non-Alcoholic Fatty Liver Disease. J Hepatol
64(6):1388–1402. https:// doi. org/ 10. 1007/ s10620- 016- 4044-2.
51. Italian Association for the Study of the Liver (AISF) (2017) AISF
position paper on nonalcoholic fatty liver disease (nafld): updates
and future directions. Dig Liver Dis 49:471–483. https:// doi. org/
10. 1016/j. dld. 2017. 01. 147
52. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella
M, Harrison SA, Brunt EM, Sanyal AJ (2018) The diagnosis
and management of nonalcoholic fatty liver disease: practice
guidance from the american association for the study of liver
diseases. Hepatology 67(1):328–357. https:// doi. org/ 10. 1002/
hep. 29367
53. Ong JP, Elariny H, Collantes R, Younoszai A, Chandhoke V,
Reines HD, Goodman Z, Younossi ZM (2005) Predictors of
nonalcoholic steatohepatitis and advanced fibrosis in morbidly
obese patients. Obes Surg 15(3):310–315. https:// doi. org/ 10.
1381/ 09608 92053 576820
54. Verma S, Jensen D, Hart J, Mohanty S (2013) Predictive value
of ALT levels for non-alcoholic steatohepatitis (NASH) and
advanced fibrosis in non-alcoholic fatty liver disease (NAFLD).
Liver Int Off J Int Assoc Study Liver. https:// doi. org/ 10. 1111/ liv.
12226
55. Lee J-H, Kim D, Kim HJ, Lee C-H, Yang JI, Kim W, Kim YJ,
Yoon J-H, Cho S-H, Sung M-W, Lee H-S (2010) Hepatic stea-
tosis index: a simple screening tool reflecting nonalcoholic fatty
liver disease. Dig Liver Dis 42(7):503–508. https:// doi. org/ 10.
1016/j. dld. 2009. 08. 002
56. Bedogni G, Bellentani S, Miglioli L, Masutti F, Passalacqua M,
Castiglione A, Tiribelli C (2006) The fatty liver index: a sim-
ple and accurate predictor of hepatic steatosis in the general
population. BMC Gastroenterol 6:33. https:// doi. org/ 10. 1186/
1471- 230X-6- 33
57. Ferraioli G (2021) Quantitative assessment of liver steato-
sis using ultrasound controlled attenuation parameter (Echo-
sens). J Med Ultrason 48:489–495. https:// doi. org/ 10. 1007/
s10396- 021- 01106-1
58. Younossi ZM, Page S, Rafiq N, Birerdinc A, Stepanova M, Hos-
sain N, Afendy A, Younoszai Z, Goodman Z, Baranova A (2011)
A Biomarker panel for non-alcoholic steatohepatitis (NASH) and
NASH-related fibrosis. Obes Surg 21(4):431–439. https:// doi.
org/ 10. 1007/ s11695- 010- 0204-1
59. Overview | Non-alcoholic fatty liver disease (NAFLD): assess-
ment and management | Guidance | NICE. https:// www. nice. org.
uk/ guida nce/ ng49 (accessed 2022–08–13).
60. Vilar-Gomez E, Chalasani N (2018) Non-invasive assessment
of non-alcoholic fatty liver disease: clinical prediction rules and
blood-based biomarkers. J Hepatol 68(2):305–315. https:// doi.
org/ 10. 1016/j. jhep. 2017. 11. 013
61. Castera L, Friedrich-Rust M, Loomba R (2019) Noninvasive
assessment of liver disease in patients with nonalcoholic fatty
liver disease. Gastroenterology 156(5):1264-1281.e4. https:// doi.
org/ 10. 1053/j. gastro. 2018. 12. 036
62. de Franchis R, Bosch J, Garcia-Tsao G, Reiberger T, Ripoll C
(2022) Baveno VII Faculty. baveno VII-renewing consensus in
portal hypertension. J Hepatol 76:959–974. https:// doi. org/ 10.
1016/j. jhep. 2021. 12. 022
63. Pons M, Augustin S, Scheiner B, Guillaume M, Rosselli M, Rod-
rigues SG, Stefanescu H, Ma MM, Mandorfer M, Mergeay-Fabre
M, Procopet B, Schwabl P, Ferlitsch A, Semmler G, Berzigotti
A, Tsochatzis E, Bureau C, Reiberger T, Bosch J, Abraldes JG,
Genescà J (2021) Noninvasive diagnosis of portal hypertension
in patients with compensated advanced chronic liver disease. Am
J Gastroenterol 116:723–732
64. Lee DH, Cho EJ, Bae JS, Lee JY, Yu SJ, Kim H, Lee KB, Han
JK, Choi BI (2021) Accuracy of two-dimensional shear wave
elastography and attenuation imaging for evaluation of patients
with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol
19(4):797-805.e7. https:// doi. org/ 10. 1016/j. cgh. 2020. 05. 034
65. Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ,
Cummings OW, Ferrell LD, Liu Y-C, Torbenson MS, Unalp-
Arida A, Yeh M, McCullough AJ, Sanyal AJ (2005) onalcoholic
steatohepatitis clinical research network. design and validation of
a histological scoring system for nonalcoholic fatty liver disease.
Hepatology 41:1313–1321. https:// doi. org/ 10. 1002/ hep. 20701
66. Lindor KD, Kowdley KV, Heathcote EJ, Harrison ME, Jorgensen
R, Angulo P, Lymp JF, Burgart L, Colin P (2004) Ursodeoxy-
cholic acid for treatment of nonalcoholic steatohepatitis: results
of a randomized trial. Hepatology 39(3):770–778. https:// doi. org/
10. 1002/ hep. 20092
67. Softic S, Gupta MK, Wang G-X, Fujisaka S, O’Neill BT, Rao
TN, Willoughby J, Harbison C, Fitzgerald K, Ilkayeva O, New-
gard CB, Cohen DE, Kahn CR (2017) Divergent effects of glu-
cose and fructose on hepatic lipogenesis and insulin signaling.
J Clin Invest 127(11):4059–4074. https:// doi. org/ 10. 1172/ JCI94
585
68. Colca JR, McDonald WG, Cavey GS, Cole SL, Holewa DD,
Brightwell-Conrad AS, Wolfe CL, Wheeler JS, Coulter KR,
Kilkuskie PM, Gracheva E, Korshunova Y, Trusgnich M, Karr
R, Wiley SE, Divakaruni AS, Murphy AN, Vigueira PA, Finck
BN, Kletzien RF (2013) Identification of a Mitochondrial target
of thiazolidinedione insulin sensitizers (mtot)–relationship to
newly identified mitochondrial pyruvate carrier proteins. PLoS
ONE 8(5):e61551. https:// doi. org/ 10. 1371/ journ al. pone. 00615 51
69. McCommis KS, Chen Z, Fu X, McDonald WG, Colca JR, Kletz-
ien RF, Burgess SC, Finck BN (2015) Loss of mitochondrial
pyruvate carrier 2 in the liver leads to defects in gluconeogen-
esis and compensation via pyruvate-alanine cycling. Cell Metab
22(4):682–694. https:// doi. org/ 10. 1016/j. cmet. 2015. 07. 028
70. Cusi K, Isaacs S, Barb D, Basu R, Caprio S, Garvey WT,
Kashyap S, Mechanick JI, Mouzaki M, Nadolsky K, Rinella ME,
Vos MB, Younossi Z (2022) American association of clinical
endocrinology clinical practice guideline for the diagnosis and
management of nonalcoholic fatty liver disease in primary care
and endocrinology clinical settings: co-sponsored by the ameri-
can association for the study of liver diseases (AASLD). Endocr
Internal and Emergency Medicine
1 3
Pract 28(5):528–562. https:// doi. org/ 10. 1016/j. eprac. 2022. 03.
010
71. European Association for the Study of the Liver (EASL); Euro-
pean Association for the Study of Diabetes (EASD); European
Association for the Study of Obesity (EASO) (2016) EASL-
EASD-EASO clinical practice guidelines for the management
of non-alcoholic fatty liver disease. J Hepatol 64(6):1388–1402.
https:// doi. org/ 10. 1016/j. jhep. 2015. 11. 004
72. Vilar-Gomez E, Calzadilla-Bertot L, Friedman SL, Gra-Oramas
B, Gonzalez-Fabian L, Lazo-Del Vallin S, Diago M, Adams LA
(2017) Serum Biomarkers can predict a change in liver fibrosis
1 year after lifestyle intervention for biopsy-proven NASH. Liver
Int 37(12):1887–1896. https:// doi. org/ 10. 1111/ liv. 13480
73. Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies
J, Balas B, Gastaldelli A, Tio F, Pulcini J, Berria R, Ma JZ,
Dwivedi S, Havranek R, Fincke C, DeFronzo R, Bannayan GA,
Schenker S, Cusi K (2006) A placebo-controlled trial of pioglita-
zone in subjects with nonalcoholic steatohepatitis. N Engl J Med
355(22):2297–2307. https:// doi. org/ 10. 1056/ NEJMo a0603 26
74. Kuchay MS, Krishan S, Mishra SK, Farooqui KJ, Singh MK,
Wasir JS, Bansal B, Kaur P, Jevalikar G, Gill HK, Choudhary
NS, Mithal A (2018) Effect of empagliflozin on liver fat in
patients with type 2 diabetes and nonalcoholic fatty liver dis-
ease: a randomized controlled trial (E-LIFT Trial). Diabetes Care
41(8):1801–1808. https:// doi. org/ 10. 2337/ dc18- 0165
75. Kitade H, Chen G, Ni Y, Ota T (2017) Nonalcoholic fatty liver
disease and insulin resistance: new insights and potential new
treatments. Nutrients 9(4):E387. https:// doi. org/ 10. 3390/ nu904
0387
76. Zelber-Sagi S, Ratziu V, Oren R (2011) Nutrition and physi-
cal activity in NAFLD: an overview of the epidemiological evi-
dence. World J Gastroenterol 17(29):3377–3389. https:// doi. org/
10. 3748/ wjg. v17. i29. 3377
77. Kugelmas M, Hill DB, Vivian B, Marsano L, McClain CJ (2003)
Cytokines and NASH: a pilot study of the effects of lifestyle
modification and vitamin E. Hepatology 38(2):413–419. https://
doi. org/ 10. 1053/ jhep. 2003. 50316
78. Shah K, Stufflebam A, Hilton TN, Sinacore DR, Klein S, Villar-
eal DT (2009) Diet and exercise interventions reduce intrahepatic
fat content and improve insulin sensitivity in obese older adults.
Obesity (Silver Spring) 17(12):2162–2168. https:// doi. org/ 10.
1038/ oby. 2009. 126
79. Ryan MC, Itsiopoulos C, Thodis T, Ward G, Trost N, Hofferberth
S, O’Dea K, Desmond PV, Johnson NA, Wilson AM (2013) The
mediterranean diet improves hepatic steatosis and insulin sen-
sitivity in individuals with non-alcoholic fatty liver disease. J
Hepatol 59(1):138–143. https:// doi. org/ 10. 1016/j. jhep. 2013. 02.
012
80. Alkhouri N, Herring R, Kabler H, Kayali Z, Hassanein T, Kohli
A, Huss RS, Zhu Y, Billin AN, Damgaard LH, Buchholtz K,
Kjær MS, Balendran C, Myers RP, Loomba R, Noureddin M
(2022) Safety and efficacy of combination therapy with semaglu-
tide, cilofexor and firsocostat in patients with non-alcoholic stea-
tohepatitis: a randomised. Open-Label Phase II Trial J Hepatol
S0168–8278(22):00235–00245. https:// doi. org/ 10. 1016/j. jhep.
2022. 04. 003
81. Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Rat-
ziu V, Sanyal AJ, Sejling A-S, Harrison SA (2021) NN9931–
4296 investigators. a placebo-controlled trial of subcutaneous
semaglutide in nonalcoholic steatohepatitis. N Engl J Med
384:1113–1124. https:// doi. org/ 10. 1056/ NEJMo a2028 395
82. Rubino DM, Greenway FL, Khalid U, O’Neil PM, Rosenstock
J, Sørrig R, Wadden TA, Wizert A, Garvey WT (2022) STEP 8
investigators. effect of weekly subcutaneous semaglutide vs daily
liraglutide on body weight in adults with overweight or obesity
without diabetes. The step 8 randomized clinical trial. JAMA
327:138–150. https:// doi. org/ 10. 1001/ jama. 2021. 23619
83. Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker
R, Hazlehurst JM, Guo K, Abouda G, Aldersley MA, Stocken
D, Gough SC, Tomlinson JW, Brown RM, Hübscher SG, New-
some PN (2016) Liraglutide safety and efficacy in patients with
non-alcoholic steatohepatitis (LEAN): a multicentre, double-
blind, randomised, placebo-controlled phase 2 study. The Lancet
387(10019):679–690. https:// doi. org/ 10. 1016/ S0140- 6736(15)
00803-X
84. A Placebo-Controlled Trial of Subcutaneous Semaglutide in
Nonalcoholic Steatohepatitis | NEJM. https://www.nejm.org/
doi/full/https:// doi. org/ 10. 1056/ NEJMo a2028 395 (accessed
2022–08–14)
85. Mayerson AB, Hundal RS, Dufour S, Lebon V, Befroy D, Cline
GW, Enocksson S, Inzucchi SE, Shulman GI, Petersen KF (2002)
The Effects of rosiglitazone on insulin sensitivity, lipolysis, and
hepatic and skeletal muscle triglyceride content in patients with
type 2 diabetes. Diabetes 51(3):797–802. https:// doi. org/ 10. 2337/
diabe tes. 51.3. 797
86. Gross B, Pawlak M, Lefebvre P, Staels B (2017) PPARs in obe-
sity-induced T2DM, dyslipidaemia and NAFLD. Nat Rev Endo-
crinol 13(1):36–49. https:// doi. org/ 10. 1038/ nrendo. 2016. 135
87. Galli A, Crabb DW, Ceni E, Salzano R, Mello T, Svegliati-Bar-
oni G, Ridolfi F, Trozzi L, Surrenti C, Casini A (2002) Antidia-
betic thiazolidinediones inhibit collagen synthesis and hepatic
stellate cell activation invivo and invitro. Gastroenterology
122(7):1924–1940. https:// doi. org/ 10. 1053/ gast. 2002. 33666
88. Musso G, Cassader M, Paschetta E, Gambino R (2017) Thiazo-
lidinediones and advanced liver fibrosis in nonalcoholic steato-
hepatitis: a meta-analysis. JAMA Intern Med 177(5):633–640.
https:// doi. org/ 10. 1001/ jamai ntern med. 2016. 9607
89. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel
S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC,
Inzucchi SE (2015) Empagliflozin, cardiovascular outcomes, and
mortality in type 2 diabetes. N Engl J Med 373(22):2117–2128.
https:// doi. org/ 10. 1056/ NEJMo a1504 720
90. Neal B, Perkovic V, Matthews DR (2017) Canagliflozin and car-
diovascular and renal events in type 2 diabetes. N Engl J Med
377(21):2099. https:// doi. org/ 10. 1056/ NEJMc 17125 72
91. Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A,
Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL,
Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson
IAM, Fredriksson M, Johansson PA, Langkilde A-M, Sabatine
MS (2019) Dapagliflozin and Cardiovascular outcomes in type 2
diabetes. N Engl J Med 380(4):347–357. https:// doi. org/ 10. 1056/
NEJMo a1812 389
92. Tahara A, Takasu T, Yokono M, Imamura M, Kurosaki E (2017)
Characterization and comparison of SGLT2 inhibitors: part 3.
Effects on diabetic complications in type 2 diabetic mice. Eur J
Pharmacol 809:163–171. https:// doi. org/ 10. 1016/j. ejphar. 2017.
05. 019
93. Gaborit B, Ancel P, Abdullah AE, Maurice F, Abdesselam I,
Calen A, Soghomonian A, Houssays M, Varlet I, Eisinger M,
Lasbleiz A, Peiretti F, Bornet CE, Lefur Y, Pini L, Rapacchi S,
Bernard M, Resseguier N, Darmon P, Kober F, Dutour A (2021)
Effect of empagliflozin on ectopic fat stores and myocardial ener-
getics in Type 2 diabetes: The EMPACEF Study. Cardiovasc Dia-
betol 20(1):57. https:// doi. org/ 10. 1186/ s12933- 021- 01237-2
94. Jain MR, Giri SR, Bhoi B, Trivedi C, Rath A, Rathod R, Ranvir
R, Kadam S, Patel H, Swain P, Roy SS, Das N, Karmakar E,
Wahli W, Patel PR (2018) Dual PPARα/γ agonist saroglitazar
improves liver histopathology and biochemistry in experimental
NASH models. Liver Int 38(6):1084–1094. https:// doi. org/ 10.
1111/ liv. 13634
Internal and Emergency Medicine
1 3
95. Inventiva’s lanifibranor meets the primary and key secondary
endpoints in the Phase IIb NATIVE clinical trial in non-alcoholic
steatohepatitis (NASH). Inventiva Pharma. https:// inven tivap
harma. com/ inven tivas- lanif ibran or- meets- the- prima ry- and-
key- secon dary- endpo ints- in- the- phase- iib- native- clini cal- trial-
in- non- alcoh olic- steat ohepa titis- nash/ (accessed 2022–08–14).
96. Ren M, Zhou X, Zhang Y, Mo F, Yang J, Yu M, Ji F (2022)
Effects of bariatric endoscopy on non-alcoholic fatty liver dis-
ease: a comprehensive systematic review and meta-analysis.
Front Endocrinol (Lausanne) 13:931519. https:// doi. org/ 10. 3389/
fendo. 2022. 931519
97. Caiazzo R, Lassailly G, Leteurtre E, Baud G, Verkindt H,
Raverdy V, Buob D, Pigeyre M, Mathurin P, Pattou F (2014)
Roux-En-Y gastric bypass versus adjustable gastric banding to
reduce nonalcoholic fatty liver disease: a 5-year controlled longi-
tudinal study. Ann Surg 260(5):893–898. https:// doi. org/ 10. 1097/
SLA. 00000 00000 000945
98. Lassailly G, Caiazzo R, Buob D, Pigeyre M, Verkindt H,
Labreuche J, Raverdy V, Leteurtre E, Dharancy S, Louvet A,
Romon M, Duhamel A, Pattou F, Mathurin P (2015) Bariatric
surgery reduces features of nonalcoholic steatohepatitis in mor-
bidly obese patients. Gastroenterology 149(2):379–388. https://
doi. org/ 10. 1053/j. gastro. 2015. 04. 014
99. Chavez-Tapia NC, Tellez-Avila FI, Barrientos-Gutierrez T,
Mendez-Sanchez N, Lizardi-Cervera J, Uribe M (2020) Bari-
atric surgery for non-alcoholic steatohepatitis in obese patients.
Cochrane Database Syst Rev. https:// doi. org/ 10. 1002/ 14651 858.
CD007 340. pub2
100. Kalinowski P, Paluszkiewicz R, Ziarkiewicz-Wróblewska B,
Wróblewski T, Remiszewski P, Grodzicki M, Krawczyk M
(2017) Liver function in patients with nonalcoholic fatty liver
disease randomized to Roux-En-Y gastric bypass versus sleeve
gastrectomy: a secondary analysis of a randomized clinical trial.
Ann Surg 266(5):738–745. https:// doi. org/ 10. 1097/ SLA. 00000
00000 002397
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
... Globally, NAFLD has reached epidemic levels, with pooled prevalence of 14% (Africa), 24% (North America) [13], 24-27% (range 18-40%, Europe) [14], 27% (Asia), 31% (South America), and 32% (Middle East) [13]. NAFLD is more frequent in men than in women (33 vs. 20%), and also affects 10-20% of the pediatric population [15]. Evidence indicates the prevalence of NAFLD is on the rise worldwide together with that of obesity and associated complications [11,16,17]. ...
... But NAFLD is no longer an isolated condition, since NAFLD is associated with morbid obesity in about 90% of the cases [59,60], with obesity and dyslipidemia in over 80% of the cases [61,62], with hypertension in 70% of cases and with type 2 diabetes (T2DM) in about 50% of the cases [63][64][65]. Such close associations strongly suggests that NAFLD is a systemic disease [10,11,15,66] increasing with poor lifestyles, and in parallel with the epidemiological raise of overweight, obesity, insulin resistance, and metabolic syndrome [4,12,[67][68][69][70]. Younossi et al. [71] studied the prevalence of NAFLD and NASH in a metanalysis including 80 studies from 20 countries worldwide and 49,419 patients with T2DM. ...
Article
Full-text available
The term non-alcoholic fatty liver disease (NAFLD) has rapidly become the most common type of chronic liver disease. NAFLD points to excessive hepatic fat storage and no evidence of secondary hepatic fat accumulation in patients with “no or little alcohol consumption”. Both the etiology and pathogenesis of NAFLD are largely unknown, and a definitive therapy is lacking. Since NAFLD is very often and closely associated with metabolic dysfunctions, a consensus process is ongoing to shift the acronym NAFLD to MAFLD, i.e., metabolic-associated fatty liver disease. The change in terminology is likely to improve the classification of affected individuals, the disease awareness, the comprehension of the terminology and pathophysiological aspects involved, and the choice of more personalized therapeutic approaches while avoiding the intrinsic stigmatization due to the term “non-alcoholic”. Even more recently, other sub-classifications have been proposed to concentrate the heterogeneous causes of fatty liver disease under one umbrella. While awaiting additional validation studies in this field, we discuss the main reasons underlying this important shift of paradigm.
... Non-alcoholic fatty liver disease (NAFLD) is becoming an epidemic all over the world, with a prevalence ranging from 14% to more than 30% [1][2][3]. NAFLD is a general term for several different liver conditions, including non-alcoholic steatohepatitis (NASH). NASH can lead to liver cirrhosis and potentially hepatocellular carcinoma (HCC) [4]. ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver condition worldwide. Numerous studies conducted recently have demonstrated a connection between the dysbiosis of the development of NAFLD and gut microbiota. Rebuilding a healthy gut ecology has been proposed as a strategy involving the use of probiotics. The purpose of this work is to investigate and compare the function of probiotics Akkermansia muciniphila (A. muciniphila) and VSL#3 in NAFLD mice. Rodent NAFLD was modeled using a methionine choline-deficient diet (MCD) with/without oral probiotic delivery. Subsequently, qPCR, histological staining, and liver function tests were conducted. Mass spectrometry-based analysis and 16S rDNA gene sequencing were used to investigate the liver metabolome and gut microbiota. We found that while both A. muciniphila and VSL#3 reduced hepatic fat content, A. muciniphila outperformed VSL#3. Furthermore, probiotic treatment restored the β diversity of the gut flora and A. muciniphila decreased the abundance of pathogenic bacteria such as Ileibacterium valens. These probiotics altered the metabolism in MCD mice, especially the glycerophospholipid metabolism. In conclusion, our findings distinguished the role of A. muciniphila and VSL#3 in NAFLD and indicated that oral-gavage probiotics remodel gut microbiota and improve metabolism, raising the possibility of using probiotics in the cure of NAFLD.
... The ability of DHA and potentially EPA to suppress adipocyte dysfunction may form the basis for the beneficial actions of these n3-PUFAs to prevent and/or reverse fatty liver disease, as we previously reported for these rats [35]. This statement relates directly to a recent publication that provides strong evidence for adipose tissue dysfunction as a causal factor in the development of NAFLD [3] and the subsequent development of insulin resistance [3,75,76]. Not only do these fatty acids decrease the size of adipocytes, they help to retain their normal functionality as indicated by reduced immune cell infiltration. ...
Article
Full-text available
Polyunsaturated fatty acids (PUFAs) can alter adipose tissue function; however, the relative effects of plant and marine n3-PUFAs are less clear. Our objective was to directly compare the n3-PUFAs, plant-based α-linolenic acid (ALA) in flaxseed oil, and marine-based eicosapentaenoic acid (EPA) or docosahexaenoic acid (DHA) in high-purity oils versus n6-PUFA containing linoleic acid (LA) for their effects on the adipose tissue and oral glucose tolerance of obese rats. Male fa/fa Zucker rats were assigned to faALA, faEPA, faDHA, and faLA groups and compared to baseline fa/fa rats (faBASE) and lean Zucker rats (lnLA). After 8 weeks, faEPA and faDHA had 11–14% lower body weight than faLA. The oral glucose tolerance and total body fat were unchanged, but faEPA had less mesenteric fat. faEPA and faDHA had fewer large adipocytes compared to faLA and faALA. EPA reduced macrophages in the adipose tissue of fa/fa rats compared to ALA and DHA, while faLA had the greatest macrophage infiltration. DHA decreased (~10-fold) T-cell infiltration compared to faBASE and faEPA, whereas faALA and faLA had an ~40% increase. The n3-PUFA diets attenuated tumour necrosis factor-α in adipose tissue compared to faBASE, while it was increased by LA in both genotypes. In conclusion, EPA and DHA target different aspects of inflammation in adipose tissue.
... For decades, type 2 diabetes (T2D) has shown close relationships with various diseases, including noncommunicable diseases (NCDs), metabolic syndrome (Met-S), dyslipidemia, fatty liver, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), and others [1]. As T2D and obesity increase, several problems of Met-S become global health and medical issues [2]. ...
Article
Full-text available
Type 2 diabetes (T2D) has close relationships with nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), and others. The European Hepatology Society International Hepatology Conference (EASL-ILC) 2023 presented the announcement of novel nomenclatures for metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). Furthermore, metabolic dysfunction-associated steatotic liver disease (MetALD) is used to describe those who consume greater amounts of alcohol, defined as more than 210g per week for males and more than 140g per week for females. MASLD may affect about one quarter of adults worldwide. Its criteria include the evidence of hepatic steatosis in three situations: obesity/overweight, T2DM, or the presence of metabolic dysfunction.
... As a metabolic organ, the liver functions in maintaining overall physiological balance. However, NAFLD could disrupt liver function, resulting in symptoms such as oxidative stress, inflammatory responses, lipid metabolism disorders, and dyslipidemia (1). Epidemiological analysis revealed NAFLD affected approximately 25% of the global population, with the incidence of nonalcoholic steatohepatitis (NASH) projected to increase by 56% by the year 2030 (2). ...
Article
Non-alcoholic fatty liver disease (NAFLD) is a prevalent liver disorder, affecting approximately 25% of the population. Coffee-drinking obese smokers exhibit lower body weights and decreased NAFLD rates, but the reasons behind this remain unclear. Additionally, the effect of nicotine, the main component of tobacco, on the development of NAFLD is still controversial. Our study aimed to explore the possible reasons that drinking coffee could alleviate NAFLD and gain weight, and identify the real role of nicotine in NAFLD of obese smokers. A NAFLD model in mice was induced by administering nicotine and a high-fat diet (HFD). We recorded changes in body weight and daily food intake, measured the weights of the liver and visceral fat, and observed liver and adipose tissue histopathology. Lipid levels, liver function, liver malondialdehyde (MDA), superoxide dismutase (SOD), serum inflammatory cytokine levels, and the expression of hepatic genes involved in lipid metabolism were determined. Our results demonstrated that nicotine exacerbated the development of NAFLD, and caffeine had a hepatoprotective effect on NAFLD. The administration of caffeine could ameliorate nicotine-plus-HFD-induced NAFLD by reducing lipid accumulation, regulating hepatic lipid metabolism, alleviating oxidative stress, attenuating inflammatory response, and restoring hepatic functions. These results might explain why obese smokers with high coffee consumption exhibit the lower incidence rate of NAFLD and tend to be leaner. It is essential to emphasize that the detrimental impact of smoking on health is multifaceted. Smoking cessation remains the sole practical and effective strategy for averting the tobacco-related complications and reducing the risk of mortality.
... This link between obesity and NAFLD is strongly backed by multiple scientific studies, which have revealed a consistent pattern of a higher Body Mass Index (BMI) and an increased waist circumference being associated with a higher prevalence of NAFLD. These studies shed light on the critical role obesity plays in exacerbating the risk of NAFLD (Machado and Cortez-Pinto, 2023;Rojano et al., 2023). ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is a prevailing health challenge that requires urgent innovative interventions. This review explores the role of nanotechnology as a promising potential in the treatment of NAFLD. It delineates the limitations of the current management strategies for NAFLD and highlights the new nanotechnology-based treatments including nanoemulsions, liposomes, micelles, polymeric nanoparticles, nanogels, inorganic nanoparticles, and zinc oxide nanoparticles. Despite the optimism surrounding the nanotechnological approach, the review underscores the need to address the limitations such as technical challenges, potential toxicity, and ethical considerations that impede the practical application of nanotechnology in NAFLD management. It advocates for collaborative efforts from researchers, clinicians, ethicists, and policymakers to achieve safe, effective, and equitable nanotechnology-based treatments for NAFLD. See also Figure 1(Fig. 1).
... Obesity is a risk factor for insulin resistance (IR) that could result in type 2 diabetes mellitus (T2DM), associated with abnormal insulin secretion and finally chronic hyperglycemia [4][5][6][7]. Moreover, a close relationship between IR and nonalcoholic fatty liver disease (NAFLD) was demonstrated, with a fivefold higher prevalence of NAFLD in patients with T2DM than in patients without T2DM [8]. ...
Article
Full-text available
Prolactin-releasing peptide (PrRP) is an anorexigenic neuropeptide that has potential for the treatment of obesity and its complications. Recently, we designed a palmitoylated PrRP31 analog (palm11-PrRP31) that is more stable than the natural peptide and able to act centrally after peripheral administration. This analog acted as an anti-obesity and glucose-lowering agent, attenuating lipogenesis in rats and mice with high-fat (HF) diet-induced obesity. In Wistar Kyoto (WKY) rats fed a HF diet for 52 weeks, we explored glucose intolerance, but also prediabetes, liver steatosis and insulin resistance-related changes, as well as neuroinflammation in the brain. A potential beneficial effect of 6 weeks of treatment with palm11-PrRP31 and liraglutide as comparator was investigated. Liver lipid profiles, as well as urinary and plasma metabolomic profiles, were measured by lipidomics and metabolomics, respectively. Old obese WKY rats showed robust glucose intolerance that was attenuated by palm11-PrRP31, but not by liraglutide treatment. On the contrary, liraglutide had a beneficial effect on insulin resistance parameters. Despite obesity and prediabetes, WKY rats did not develop steatosis owing to HF diet feeding, even though liver lipogenesis was enhanced. Plasma triglycerides and cholesterol were not increased by HFD feeding, which points to unincreased lipid transport from the liver. The liver lipid profile was significantly altered by a HF diet that remained unaffected by palm11-PrRP31 or liraglutide treatment. The HF-diet-fed WKY rats revealed astrogliosis in the brain cortex and hippocampus, which was attenuated by treatment. In conclusion, this study suggested multiple beneficial anti-obesity-related effects of palm11-PrRP31 and liraglutide in both the periphery and brain.
Article
Obesity is a multifactorial, chronic, progressive and recurrent disease considered a public health issue worldwide and an important determinant of disability and death. In Spain, its current prevalence in the adult population is about 24% and an estimated prevalence in 2035 of 37%. Obesity increases the probability of several diseases linked to higher mortality such as diabetes, cardiovascular disease, hyperlipidemia, arterial hypertension, non-alcoholic fatty liver disease, several types of cancer, or obstructive sleep apnea. On the other hand, although the incidence of inflammatory bowel disease (IBD) is stabilizing in Western countries, its prevalence already exceeds 0.3%. Paralleling to general population, the current prevalence of obesity in adult patients with IBD is estimated at 15-40%. Obesity in patients with IBD could entail, in addition to its already known impact on disability and mortality, a worse evolution of the IBD itself and a worse response to treatments. The aim of this document, performed in collaboration by four scientific societies involved in the clinical care of severe obesity and IBD, is to establish clear and concise recommendations on the therapeutic possibilities of severe or typeIII obesity in patients with IBD. The document establishes general recommendations on dietary, pharmacological, endoscopic, and surgical treatment of severe obesity in patients with IBD, as well as pre- and post-treatment evaluation.
Article
Full-text available
Background and objectiveEndoscopic bariatric and metabolic therapies (EBMTs) are emerging minimally invasive therapeutic options for obesity and its related complications, including non-alcoholic fatty liver disease (NAFLD). This study aimed to evaluate the effects of EBMTs on NALFD in patients with obesity.Methods Four databases were searched until Nov 2021. Randomized controlled trials (RCTs) and observational studies reporting liver-related outcomes following Food and Drug Administration (FDA)-approved and non-FDA-approved EBMTs were included. Liver parameters, metabolic parameters, and weight loss were evaluated. Risk of bias was assessed using the “risk of bias” tool in the Cochrane Collaboration for RCTs and the Methodological Index for Non-Randomized Studies criteria for observational studies.ResultsThirty-three studies with 1710 individuals were included. Regarding the effects of EBMTs on liver fibrosis, a significant decline of NAFLD Fibrosis Score, but not transient elastography-detected liver stiffness or Fibrosis-4 Index, was observed. EBMTs significantly improved liver steatosis (control attenuation parameter and Hepatic Steatosis Index), NAFLD Activity Score, and Homeostasis Model Assessment of Insulin Resistance. EBMTs reduced serum levels of alanine transaminase, aspartate aminotransferase, and gamma-glutamyl transpeptidase considerably. Moreover, EBMTs had reducing effects on the serum levels of triglycerides and total cholesterol as well as body weight.Conclusions Our meta-analysis suggested that EBMTs could ameliorate NAFLD based on the evidence of improved liver steatosis, liver function, and insulin resistance. Large-scale, prospective, long-term studies are warranted to clarify the role of EBMTs in patients with different stages of NAFLD.
Article
Full-text available
Nonalcoholic steatohepatitis (NASH) is a leading cause of cirrhosis in western countries. Insulin resistance (IR), type 2 diabetes (T2D), and the polymorphisms patatin-like phospholipase domain-containing 3 (PNPLA3) rs738409 and transmembrane 6 superfamily member 2 (TM6SF2) rs58542926 are independent risk factors of NASH. Nevertheless, little is known about the interaction between IR and T2D with these polymorphisms in the pathogenesis of NASH and the development of advanced fibrosis. Thus, our study aimed to investigate this relationship. This is a cross-sectional study including NASH patients diagnosed by liver biopsy, at the Vall d'Hebron University Hospital. A total of 140 patients were included (93 T2D, 47 non-T2D). T2D (OR = 4.67; 95%CI 2.13-10.20; p < 0.001), PNPLA3 rs738409 and TM6SF2 rs58542926 polymorphisms (OR = 3.94; 95%CI 1.63-9.54; p = 0.002) were independently related with advanced liver fibrosis. T2D increased the risk of advance fibrosis on top of the two polymorphisms (OR = 14.69; 95%CI 3.03-77.35; p = 0.001 for PNPLA3 rs738409 and OR = 11.45; 95%CI 3.16-41.55; p < 0.001 for TM6SF2 rs58542926). In non-T2D patients, the IR (HOMA-IR ≥ 5.2, OR = 14.33; 95%CI 2.14-18.66; p = 0.014) increased the risk of advanced fibrosis when the polymorphisms were present (OR = 19.04; 95%CI 1.71-650.84; p = 0.042). The T2D and IR status increase the risk of advanced fibrosis in patients with NASH carrying the PNPLA3 rs738409 and/or TM6SF2 rs58542926 polymorphisms, respectively.
Article
Full-text available
Importance Phase 3 trials have not compared semaglutide and liraglutide, glucagon-like peptide-1 analogues available for weight management. Objective To compare the efficacy and adverse event profiles of once-weekly subcutaneous semaglutide, 2.4 mg, vs once-daily subcutaneous liraglutide, 3.0 mg (both with diet and physical activity), in people with overweight or obesity. Design, Setting, and Participants Randomized, open-label, 68-week, phase 3b trial conducted at 19 US sites from September 2019 (enrollment: September 11-November 26) to May 2021 (end of follow-up: May 11) in adults with body mass index of 30 or greater or 27 or greater with 1 or more weight-related comorbidities, without diabetes (N = 338). Interventions Participants were randomized (3:1:3:1) to receive once-weekly subcutaneous semaglutide, 2.4 mg (16-week escalation; n = 126), or matching placebo, or once-daily subcutaneous liraglutide, 3.0 mg (4-week escalation; n = 127), or matching placebo, plus diet and physical activity. Participants unable to tolerate 2.4 mg of semaglutide could receive 1.7 mg; participants unable to tolerate 3.0 mg of liraglutide discontinued treatment and could restart the 4-week titration. Placebo groups were pooled (n = 85). Main Outcomes and Measures The primary end point was percentage change in body weight, and confirmatory secondary end points were achievement of 10% or more, 15% or more, and 20% or more weight loss, assessed for semaglutide vs liraglutide at week 68. Semaglutide vs liraglutide comparisons were open-label, with active treatment groups double-blinded against matched placebo groups. Comparisons of active treatments vs pooled placebo were supportive secondary end points. Results Of 338 randomized participants (mean [SD] age, 49 [13] years; 265 women [78.4%]; mean [SD] body weight, 104.5 [23.8] kg; mean [SD] body mass index, 37.5 [6.8]), 319 (94.4%) completed the trial, and 271 (80.2%) completed treatment. The mean weight change from baseline was –15.8% with semaglutide vs –6.4% with liraglutide (difference, –9.4 percentage points [95% CI, –12.0 to –6.8]; P < .001); weight change with pooled placebo was –1.9%. Participants had significantly greater odds of achieving 10% or more, 15% or more, and 20% or more weight loss with semaglutide vs liraglutide (70.9% of participants vs 25.6% [odds ratio, 6.3 {95% CI, 3.5 to 11.2}], 55.6% vs 12.0% [odds ratio, 7.9 {95% CI, 4.1 to 15.4}], and 38.5% vs 6.0% [odds ratio, 8.2 {95% CI, 3.5 to 19.1}], respectively; all P < .001). Proportions of participants discontinuing treatment for any reason were 13.5% with semaglutide and 27.6% with liraglutide. Gastrointestinal adverse events were reported by 84.1% with semaglutide and 82.7% with liraglutide. Conclusions and Relevance Among adults with overweight or obesity without diabetes, once-weekly subcutaneous semaglutide compared with once-daily subcutaneous liraglutide, added to counseling for diet and physical activity, resulted in significantly greater weight loss at 68 weeks. Trial Registration ClinicalTrials.gov Identifier: NCT04074161
Article
Full-text available
Controlled attenuation parameter (CAP) is the algorithm available on the FibroScan system (Echosens, France) for quantification of liver steatosis. It assesses the ultrasound beam attenuation, which is directly related to liver fat content. The inter-observer reproducibility of the technique is high, with a reported concordance correlation coefficient of 0.82. Specific quality criteria for CAP measurements are not clearly defined yet, and there are conflicting results in the literature. Using liver biopsy as the reference standard, several studies have assessed the CAP performance in grading liver steatosis, and have reported that values are not affected by liver fibrosis. The cutoff for detection of liver steatosis reported in the literature ranges from 222 decibels per meter (dB/m) in a cohort of patients with chronic hepatitis C to 294 dB/m in a meta-analysis of nonalcoholic fatty liver disease (NAFLD) patients. CAP has been used as a tool to noninvasively evaluate the prevalence of NAFLD in groups at risk or in the general population; however, it should be underscored that different CAP cutoffs for steatosis detection (S > 0) were used in different studies, and this limits the robustness of the findings. CAP, alone or combined with other noninvasive indices or biomarkers, has been proposed as a tool for assessing nonalcoholic steatohepatitis or as a noninvasive predictor of prognosis in patients with chronic liver disease. CAP is easy to perform and has become a point-of-care technique. However, there is a large overlap of values between consecutive grades of liver steatosis, and cutoffs are not clearly defined.
Article
Full-text available
Background Non-alcoholic fatty liver disease, or as recently proposed ‘metabolic-associated fatty liver disease’ (MAFLD), is characterized by pathological accumulation of triglycerides and other lipids in hepatocytes. This common disease can progress from simple steatosis to steatohepatitis, and eventually end-stage liver diseases. MAFLD is closely related to disturbances in systemic energy metabolism, including insulin resistance and atherogenic dyslipidemia. Scope of review The liver is the central organ in lipid metabolism by secreting very low density lipoproteins (VLDL) and, on the other hand, by internalizing fatty acids and lipoproteins. This review article discusses recent research addressing hepatic lipid synthesis, VLDL production and lipoprotein internalization as well as the lipid exchange between adipose tissue and liver in the context of MAFLD. Major conclusions Liver steatosis in MAFLD is triggered by excessive hepatic triglyceride synthesis utilizing fatty acids derived from white adipose tissue (WAT), de novo lipogenesis (DNL) and endocytosed remnants of triglyceride-rich lipoproteins. In consequence of high hepatic lipid content VLDL secretion is enhanced, which is the primary cause of complex dyslipidemia typical for subjects with MAFLD. Interventions reducing VLDL secretory capacity attenuate dyslipidemia while they exacerbate MAFLD, indicating that the balance of lipid storage versus secretion in hepatocytes is a critical parameter determining disease outcome. Proof of concept studies have shown that promoting lipid storage and energy combustion in adipose tissues reduces hepatic lipid load and thus ameliorates MAFLD. Moreover, hepatocellular triglyceride synthesis from DNL and WAT-derived fatty acids can be targeted to treat MAFLD. However, more research is needed to understand how individual transporters, enzymes and their isoforms affect steatosis and dyslipidemia in vivo, and whether these two aspects of MAFLD can be selectively treated. Processing of cholesterol-enriched lipoproteins appears less important for steatosis. It may, however, modulate inflammation and consequently MAFLD progression.
Article
Objective To provide evidence-based recommendations regarding the diagnosis and management of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) to endocrinologists, primary care clinicians, health care professionals, and other stakeholders. Methods The American Association of Clinical Endocrinology conducted literature searches for relevant articles published from January 1, 2010, to November 15, 2021. A task force of medical experts developed evidence-based guideline recommendations based on a review of clinical evidence, expertise, and informal consensus, according to established American Association of Clinical Endocrinology protocol for guideline development. Recommendation Summary This guideline includes 34 evidence-based clinical practice recommendations for the diagnosis and management of persons with NAFLD and/or NASH and contains 385 citations that inform the evidence base. Conclusion NAFLD is a major public health problem that will only worsen in the future, as it is closely linked to the epidemics of obesity and type 2 diabetes mellitus. Given this link, endocrinologists and primary care physicians are in an ideal position to identify persons at risk on to prevent the development of cirrhosis and comorbidities. While no U.S. Food and Drug Administration-approved medications to treat NAFLD are currently available, management can include lifestyle changes that promote an energy deficit leading to weight loss; consideration of weight loss medications, particularly glucagon-like peptide-1 receptor agonists; and bariatric surgery, for persons who have obesity, as well as some diabetes medications, such as pioglitazone and glucagon-like peptide-1 receptor agonists, for those with type 2 diabetes mellitus and NASH. Management should also promote cardiometabolic health and reduce the increased cardiovascular risk associated with this complex disease.
Article
Background & aims Non-alcoholic steatohepatitis (NASH) is associated with increased risk of liver-related and cardiovascular morbidity and mortality. Given the complex pathophysiology of NASH, combining therapies with complementary mechanisms may be beneficial. This trial evaluated the safety and efficacy of semaglutide, a glucagon-like peptide-1 receptor agonist, alone and in combination with the farnesoid X receptor agonist cilofexor and/or the acetyl-coenzyme A carboxylase inhibitor firsocostat in patients with NASH. Methods This was a phase II, open-label, proof-of-concept trial in which patients with NASH (F2–F3 on biopsy, or magnetic resonance imaging proton density fat fraction (MRI-PDFF) ≥10% and liver stiffness by transient elastography [≥7 kPa]) were randomised to 24 weeks’ treatment with semaglutide 2.4 mg once-weekly as monotherapy or combined with once-daily cilofexor (30 or 100 mg) and/or once-daily firsocostat 20 mg. The primary endpoint was safety. All efficacy endpoints were exploratory. Results A total of 108 patients were randomised to semaglutide (n=21), semaglutide plus cilofexor 30 mg (n=22), semaglutide plus cilofexor 100 mg (n=22), semaglutide plus firsocostat (n=22) or semaglutide, cilofexor 30 mg and firsocostat (n=21). Treatments were well tolerated, with incidence of adverse events similar across groups (73–90%) and most events gastrointestinal. Despite similar weight loss (7–10%), compared with semaglutide monotherapy, combinations resulted in greater improvements in liver steatosis measured by MRI-PDFF (least-squares mean of absolute changes: −9.8 to −11.0% vs. −8.0%), liver biochemistry, and noninvasive tests of fibrosis. Conclusions In patients with mild-to-moderate fibrosis due to NASH, semaglutide with firsocostat and/or cilofexor was generally well tolerated. In exploratory efficacy analyses, treatment resulted in additional improvements in liver steatosis and biochemistry vs. semaglutide alone. Given this was a small-scale open-label trial, double-blind placebo-controlled trials with adequate patient numbers to assess efficacy and safety of these combinations in NASH are warranted. Lay summary Nonalcoholic fatty liver disease (NAFLD) and its more severe form, nonalcoholic steatohepatitis (NASH), are serious liver conditions that worsen over time if untreated. The reasons people develop NASH are complex and combining therapies that target different aspects of the disease may be more helpful than using single treatments. This trial showed that the use of three different types of drugs, namely semaglutide, cilofexor and firsocostat, in combination was safe and may offer additional benefits over treatment with semaglutide alone. Trial registration number NCT03987074
Article
To expand on the work of previous meetings, a virtual Baveno VII workshop was organised for October 2021. Among patients with compensated cirrhosis or compensated advanced chronic liver disease (cACLD - defined at the Baveno VI conference), the presence or absence of clinically significant portal hypertension (CSPH) is associated with differing outcomes, including risk of death, and different diagnostic and therapeutic needs. Accordingly, the Baveno VII workshop was entitled "Personalized Care for Portal Hypertension". The main fields of discussion were the relevance and indications for measuring the hepatic venous pressure gradient as a gold standard, the use of non-invasive tools for the diagnosis of cACLD and CSPH, the impact of aetiological and non-aetiological therapies on the course of cirrhosis, the prevention of the first episode of decompensation, the management of an acute bleeding episode, the prevention of further decompensation, as well as the diagnosis and management of splanchnic vein thrombosis and other vascular disorders of the liver. For each of these 9 topics, a thorough review of the medical literature was performed, and a series of consensus statements/recommendations were discussed and agreed upon. A summary of the most important conclusions/recommendations derived from the workshop is reported here. The statements are classified as unchanged, changed, and new in relation to Baveno VI.
Article
Adiposity-Based Chronic Disease (ABCD) is a chronic disease and requires life-long treatment and follow-up. Obesity protects obesity through altered regulation of caloric intake and set point mechanisms that maintains a high equilibrium body weight. Lifestyle interventions and obesity medications do not permanently alter the set point which often makes weight loss achieved by lifestyle short-lived and operates to drive weight regain once medications are discontinued. Bariatric surgery procedures can alter appetite and lower the “set point” for equilibrium body weight via unknown mechanisms. However, few patients attain ideal body weight following surgery, many regain weight, and all require long-term follow-up for the disease. The excess adiposity of ABCD gives rise to complications that impair health and confer morbidity and mortality; however, the genetic risks and potential interactions between genes and environment that give rise to complications also cannot be eliminated. The equilibrium body weight around which set point mechanisms operate can be modified by environment, which underscores the importance of a less obesogenic environment for prevention and treatment of ABCD on a population basis. If ABCD will eventually be curable, this will depend on a clear understanding of the molecular mechanisms that determine the set point regulation of body weight, and an ability to permanently modulate the set point to oscillate around and a lean body mass. The conceptualization of ABCD as a chronic disease, however, does present us with opportunities for primary, secondary, and tertiary prevention to avert disease progression. For tertiary care, the advent of new, more effective, second-generation obesity medications will allow clinicians to treat-to-target via active management of body weight into a target range that will ameliorate specific complications.
Article
Non-alcoholic fatty liver disease (NAFLD) is a public health problem worldwide. This narrative Review provides an overview of the current literature to support the notion that NAFLD is a multisystem disease. Convincing evidence shows a strong association between NAFLD and the risk of developing multiple extrahepatic complications such as type 2 diabetes, cardiovascular disease (ie, the predominant cause of mortality in people with NAFLD), chronic kidney disease, and some types of extrahepatic malignancies. The magnitude of this risk parallels the severity of NAFLD (especially the stage of liver fibrosis). There are probably multiple underlying mechanisms by which NAFLD might increase the risk of cardiovascular disease, type 2 diabetes, and extrahepatic complications. Addressing the growing burden of NAFLD will require setting up a multidisciplinary working group and framework to progress and embrace novel collaborative ways of working to deliver holistic, person-centred care and management of people with NAFLD.