Skip to main content
Erschienen in: Internal and Emergency Medicine 4/2023

Open Access 17.02.2023 | IM - REVIEW

NAFLD, MAFLD, and beyond: one or several acronyms for better comprehension and patient care

verfasst von: Piero Portincasa

Erschienen in: Internal and Emergency Medicine | Ausgabe 4/2023

Abstract

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.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s11739-023-03203-0.
The original online version of this article was revised: In this article the word painless was changed as painful under the section NAFLD and metabolic dysfunctions. also, a grammatical change in text under Conclusions and future perspectives section has been amended.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s11739-023-03252-5.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The acronyms NASH (non-alcoholic steatohepatitis) and NAFLD (non-alcoholic fatty liver disease) were originally coined by Ludwig et al. in 1980 [1] and Shaffner and Thaler in 1986 [2], respectively. NAFLD is a clinico-histopathologic entity defining excessive hepatic fat storage without evidence of secondary hepatic fat accumulation [37] in patients with “no or little alcohol consumption”. Hepatic fat content must be greater than 5% at liver histology [8] or 5.6% at magnetic resonance [9].
Since 2020, a debate is taking place worldwide to redefine NAFLD as metabolic (dysfunction)-associated fatty liver disease (MAFLD), according to the most prevalent causes of liver steatosis [1012]. Meantime, further terminologies are being proposed instead of NAFLD. While awaiting additional validation studies, the main reasons underlying this important shift of paradigm are discussed in the following paragraphs.

NAFLD and the burden of disease: prevalence, natural history, etiology and pathogenesis

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].
In this scenario, we can expect a time-dependent increase of the incidence of liver fibrosis, decompensated liver cirrhosis, hepatocellular carcinoma, and liver-related mortality due to the progressive deterioration of initial NAFLD in the affected population [17].
The natural history of NAFLD can vary, with most patients developing a benign or slowly progressive form, which is usually asymptomatic in the early stages. The spectrum ranges from simple steatosis (non-alcoholic fatty liver, NAFL) to the progressive non-alcoholic steatohepatitis (NASH) in about 20% of cases [18]. NASH has the potential to progress to compensated and decompensated liver cirrhosis and, with more than a tenfold increase in risk, to hepatocellular carcinoma (HCC) (Fig. 1) [9, 19]. Of note, NAFLD is also a risk factor for several extrahepatic, especially metabolic manifestations [20] and is linked with increased cardiovascular risk [21].
Excessive liver fat storage has a series of well-known causes (Supplementary Table 1) and originates from a dynamic balance between causal and protective factors (Fig. 1) [22, 23].
In this scenario, individual lifestyle changes and environmental factors can promote epigenetic mechanisms such as histone methylation, abnormal DNA methylation, miRNA profiles, able to affect gene expression and to influence the progression of disease. Examples are the consequences of metabolic disturbances in pregnancy on NAFLD offspring [24], and the pro-inflammatory liver response and weight gain in germ-free mice colonized with stool microbes from 2-week-old infants born to obese mothers [25]. These mechanisms interact with inherited risk factors and modulate individual susceptibility to NAFLD [26].
Lifestyle and dietary habits [27, 28] play a key role, and NAFLD is commonly associated with metabolic abnormalities [9] such as obesity, type 2 diabetes, dyslipidemia, hypertension, hypopituitarism, and sedentary life [20]. On the other hand, alcohol, air pollution [29, 30], food contaminants [31], and gut dysbiosis, e.g., higher proportion of Proteobacteria and E. coli, with a lower proportion of Firmicutes, especially F. prausnitzii [32] likely contribute to the onset and progression of NAFLD [20, 33, 34].
In the context of metabolic abnormalities, several pathogenic pathways are involved in lipotoxicity and can contribute to the onset and progression of NAFLD, according to the nomenclature in use. The “lean” healthy visceral adipose tissue expresses anti-inflammatory cytokines (i.e., adiponectin, interleukin IL-4, IL-10, IL-13, transforming growth factor (TGF)-β, and nitric oxide (NO)) which control M2 macrophagic response and inhibit the neutrophil-mediated inflammation. By contrast, during expansion of hypertrophic (and apoptotic) visceral adipose tissue, secretion of pro-inflammatory molecules such as leptin, resistin, IL-6 and tumor necrosis factor (TNF)-α occurs. This step activates a M1 macrophage response [35], and results in insulin resistance, a chronic “metabolic” inflammatory status, and increased lipolysis of triglycerides with abundant flux of blood long-chain free fatty acids (FFA) to the liver. In addition, FFA in blood increases because of fat-enriched dietary habits and from dietary sugars driving the de novo lipogenesis (DNL) in the liver. Altogether, these factors contribute to the expansion of the hepatocyte FFA pool which can stress the mitochondrial β-oxidation capacity, lead to defective secretion/export of very-low density lipoproteins (VLDL) to blood. Accumulation of lipotoxic species such as lysophosphatidylcholine, diacylglycerol, and ceramides mediates the endoplasmic reticulum (ER) stress, the cellular oxidative stress, and the activation of the inflammasome. This is a component of the innate immunity response consisting of a multiprotein cytoplasmic complex activated by several damage-associated molecular patterns (DAMPs). Additional abnormalities consist of dysregulation of adipocytokines, depleted mitochondrial ATP, production of toxic uric acid, periodic hypoxia (i.e., during sleep apnea in extremely obese patients), and toxic products from gut microbiome which include tumor necrosis factor (TNF)-α, endogenous ethanol, and endotoxins like lipopolysaccharides (LPS). Studies in pure fatty liver models without fibrosis suggest that lipid accumulation developing with obesity can induce a distortion of liver architecture manifesting with reduced sinusoidal space and increased intrahepatic vascular resistance. Such changes can pave the way to portal hypertension observed in obesity, and progression to hemodynamically decompensated liver cirrhosis [3639].
Conditions can promote the NASH phenotype which manifests with hepatocellular injury, inflammation, stellate cell activation and progressive accumulation of excess extracellular matrix. Additional targets of the ongoing cellular damage include intracellular organelles, the nucleus, receptors and signaling pathways [4044].

NAFLD and metabolic dysfunctions

As mentioned earlier, the diagnosis of NAFLD is based on hepatic steatosis at imaging techniques or histology, and “exclusion” of competing causes of liver disease including “significant” alcohol intake. Such strict definition has some limitations. Despite the NAFLD prognosis depends on the presence of fibrotic NASH, the ultimate utility of liver biopsy becomes questionable [45]. In fact, liver biopsy is invasive, painful, risky, prone to misclassification due to sampling errors, not easily performed in large groups of patients who still lack targeted therapies for NAFLD. The issue of alcohol intake in NAFLD deserves additional observations. A standard drink contains about 14 g of pure alcohol (Rethinking Drinking Homepage—NIAAA (nih.gov)) and the current definition for NAFLD must exclude a weekly intake of ≥ 21 and ≥ 14 drinks in males and females, respectively. Above this cutoff value, the risk of alcoholic fatty liver disease (ALD) increases [46, 47], but it is difficult to exactly calculate the intake of alcoholic units or the duration of alcohol abstinence. Phosphatidylethanol can become a potential biomarker of alcohol consumption [48]. Yet, the effect of modest alcohol intake at lower cutoff values is still controversial in NAFLD individuals [49, 50].
In addition, the diagnosis of NAFLD requires the exclusion of several other causes of liver steatosis such as viral hepatitis [5154], hepatotoxic drugs [55], Wilson’s disease [56], total parenteral nutrition, prolonged fasting [57], and several other less common conditions (Supplementary Table 1). Both NAFLD and ALD rank as the most frequent conditions [58]. On one hand, the diagnosis of NAFLD relies on exclusion criteria and does not require the presence of metabolic dysfunction. 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 [6365]. 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, 6770]. 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. The prevalence of NAFLD was more than twofold higher than in the general population, i.e., 56%. In the same group, the prevalence of NASH was 37%. In patients with NAFLD and T2DM undergoing liver biopsy, 17% had advanced (F3–F4) liver fibrosis [71]. The close association of NAFLD with systemic metabolic conditions also explains why cardiovascular complications are frequent in NAFLD patients [65, 72], including atrial fibrillation [73, 74], diabetes, chronic kidney disease and extrahepatic neoplasms [7577]. It is worth noting that 10–30% of non-obese individuals can have NAFLD [4, 78].
Another close link between NAFLD and metabolic dysfunction is that the metabolic syndrome is often associated with NAFLD and with increased cardiovascular disease. NAFLD per se, however, is independently associated with cardiovascular disease [65, 72, 79, 80].
The ultimate knowledge of complex mechanisms governing the onset and progression of NAFLD is poorly known. The term “steatosis” is intrinsically characterized by the interplay of multifactorial factors [34, 40, 81, 82]. The coexistence of such factors makes the diagnosis of NAFLD and the design of clinical trials often difficult, since several confounding factors can be present [9].

MAFLD: debate about nomenclature

The acronym NAFLD was originally established as a diagnosis of exclusion based on the use of the stigmatizing term “alcoholic” and on a poor pathophysiological knowledge. In the past years, this term has generated confusion or uncertainty with regard to knowledge and allocation in clinical trials [83, 84]. Studies show that up to 96% of subjects with NAFLD can be unaware they have liver disease [85].
Following the current directions, there is a need for adopting a better term for liver steatosis to emphasize what the disease “is”, rather than what “it is not” when considering the burden of contributing metabolic abnormalities, pathophysiological mechanisms, diagnostic, and therapeutic strategies [84]. Since 2020 experts from 134 countries have proposed the transition of terminology from NAFLD to metabolic dysfunction-associated fatty liver disease (MAFLD) [10] which points to the close association between fatty liver, metabolic disorders and target organ dysfunction (i.e., diabetes, chronic kidney disease, atherosclerosis, lung dysfunction, colon cancer, and both intrahepatic and extrahepatic events) [86], rather than on exclusion criteria [12]. The new terminology is not yet endorsed by international societies, including European Association for the Study of Liver Disease (EASL) and the American Association for the Study of Liver Disease AASLD) [87]. MAFLD is based on the presence of fatty liver at imaging, or by the combination of serum biomarkers or liver biopsy (Table 1) in individuals with overweight/obesity (different in Caucasians or Asians), T2DM (i.e., HbA1c ≥ 6.5% or specific drug treatment). In lean/normal weight individuals, the diagnosis is based on the presence of metabolic dysregulation with at least two out of seven abnormalities which include increased waist circumference, blood pressure, hypertriglyceridemia, low plasma high-density lipoprotein cholesterol, impaired fasting plasma glucose, insulin resistance with impaired Homeostatic Model Assessment of Insulin Resistance, and subclinical inflammation by plasma high‐sensitivity C‐reactive protein level [10, 83, 8892] (Fig. 2A).
Table 1
Diagnosis of liver steatosis/fibrosis/inflammation/progression
Outcome
Methods
 
Notes
Steatosis
Serum biomarkers
AST, ALT, GGT
Not specific, can fluctuate or remain normal during progression of disease
 
Algorithms
Hepatic Steatosis index
Fatty Liver Index
Combined serum tests and metabolic information [110, 129]
 
Imaging
Abdominal ultrasound
Easily available, noninvasive
Low sensitivity for steatosis degree < 30%
  
ATI
To be validated [130]
  
MRI-PDFF
Expensive, limited availability
 
CAP
Software (component of transient elastography)
Quantitative measurement of liver steatosis [131]
Fibrosis
Serum biomarkers
FIB-4, NFS, ELF
Combined anthropometric data and serum tests. Can exclude advanced fibrosis [132]
 
Imaging
TE
Validated, accurate, associated with measurement of liver stiffness and steatosis (CAP). Decreased sensitivity in obese patients [133]
  
2D-SWE
Liver stiffness
  
MRI-MRE
Expensive, limited availability
Steatohepatitis
Imaging
SWDS
To be validated [130]
Progression of disease
Liver biopsy
Steatosis, fibrosis, inflammation
Invasive, limitations (see paragraph)
ALT alanine aminotransferase, AST aspartate aminotransferase, ATI attenuation imaging, CAP controlled attenuation parameter, 2D-SWE two-dimensional shear wave elastography, ELF enhanced liver fibrosis test, GGT γ-glutamyl transferase, MRE magnetic resonance elastography, MRI magnetic resonance imaging, NFS NAFLD fibrosis score, PDFF proton density fat fraction, SWDS shear wave dispersion slope (SWDS), TE transient elastography
According to this perspective, the adoption of MAFLD definition provides a better classification of patients at higher risk of cardiovascular and kidney diseases [21, 9395], and hepatic fibrosis [9698], independently of other causes of liver damage. For example, MAFLD terminology becomes independent of alcohol intake, a situation which appears to worsen hepatic fibrosis and increase mortality in liver steatosis [99103]. On the other hand, one could argue that liver disease in general is often associated with alcohol abuse in the perception of the general public, and that adding “non-alcoholic” would be beneficial since it is now explicitly stated that the liver disease is not related to alcohol.
The presence of viral hepatitis is not an “a priori” exclusion criterium for MAFLD. In addition, MAFLD represents a further risk factor for HCC in hepatitis C virus (HCV) and hepatitis B virus (HBV) patients [104, 105]. Vice versa, the 10-year risk of cardiovascular disease is higher in patients with MAFLD and concomitant viral infection by hepatitis B or C virus, than in patients with only MAFLD [106]. With MAFLD, moreover, the role of metabolic abnormalities on liver damage will be elaborated together with other causes of liver steatosis, such as drugs, pregnancy and gut surgery [107109]. This possibility will be labeled as “MAFLD plus additional cause ….” [11, 96].
The diagnosis of MAFLD can include the presence of positive biomarkers independently from imaging and histology. This is not the case in NAFLD (see above). Thus, the possibility exists that a patient referred with a high-score for a specific algorithm such as the fatty liver index [110]) will be classified as MAFLD in the presence of metabolic dysfunction.
By contrast, the criteria designed for MAFLD do not extend to individuals with liver steatosis without metabolic dysfunction.
In a recent metanalysis and systematic review involving a pool of over 3 million individuals, the prevalence of MAFLD was 39%, 30%, and 5% in obese, non-obese, and normal weight individuals, respectively. Although not all cases of NAFLD are MAFLD [93, 111], by adopting the MAFLD terminology, the clinicians can better understand the pathophysiological mechanisms of disease. In terms of risk assessment, although evidence is limited, MAFLD (but not pure NAFLD) can be directly related with all-cause mortality [112, 113]. NAFLD has been linked with the development of cardiovascular diseases [114], although this association seems mainly linked with the metabolic components. In fact, NAFLD subjects not classifiable as MAFLD are at lower risk [21, 115]. Therapeutic approaches will improve since MAFLD points to the interaction of several pathophysiological factors and the multidisciplinary collaboration between internists, cardiologists, endocrinologists, nutritionists, hepatologists, and family medicine [10, 11, 83, 91, 94, 115122]. In addition, the target populations will gain a better comprehension of terminology [88, 123] without feeling stigmatized because of the word “alcoholic” [12, 119, 121, 124].
The change in terminology, along with either exclusion or inclusion criteria for NAFLD and MAFLD, respectively, creates three groups partially overlapping (Fig. 2B):
  • “Pure” NAFLD (non-MAFLD) where metabolic dysfunction is absent, and significant alcohol intake is excluded.
  • “Overlapping” N(M)AFLD where metabolic dysfunction is present and significant alcohol intake is excluded.
  • “Pure” MAFLD (non-NAFLD) where metabolic dysfunction is present and alcohol consumption can be significant.
Furthermore, in terms of risk assessment related to metabolic phenotypes (all-cause mortality risk, cardiovascular risk, histological progression of liver disease), subjects with overlapping or pure MAFLD represent a heterogeneous group. We speculate that further studies should assess the suitability of a more detailed risk stratification of MAFLD based on specific metabolic phenotypes (Table 2). Recent evidence in a large Korean cohort reported a higher cardiovascular disease risk in lean MAFLD or MAFLD associated with diabetes mellitus, than in overweigh MAFLD subjects, irrespective of metabolic abnormalities or comorbidities. In this cohort, the cardiovascular risk was linked with advanced liver fibrosis irrespective of MAFLD subtype [125].
Table 2
Possible sub-classification of subjects with MAFLD according to metabolic phenotype
1. Normal weight, metabolic unhealthy, without type 2 diabetes
2. Normal weight, metabolic unhealthy, with type 2 diabetes
3. Overweight, metabolic unhealthy, without type 2 diabetes
4. Overweight, metabolic unhealthy, with type 2 diabetes
5. Obese, without type 2 diabetes
6. Obese, with type 2 diabetes
It is evident that further studies are required to better focus on risk-specific sub-profiles (adjusted for confounders) and natural history of MAFLD/NAFLD association between MAFLD and hypertension and diabetes in the last two groups [126]. Despite few studies have dealt with the comparison between NAFLD and MAFLD, we still need caution due to the nature of the studies (retrospective), selection of groups, statistical issues and conclusions of the studies [96, 127].

Fatty liver disease: more acronyms on the way

Notably, a further classification of liver steatosis is being proposed under the general umbrella of fatty liver disease (FLD). Sub-classifications include almost all possible combinations of genetic, lipodystrophy, metabolic, alcoholic, combined, and yet-to-be-defined causes. This schematic classification will stimulate a further discussion with the aim to improve both comprehension and diagnostic/therapeutic approaches for FLD populations (Table 3) (Fig. 3) [84]. The discussion has gone further with a novel taxonomic classification of NAFLD based on hepatic, pathogenic and systemic features of disease in the individual patient [86, 128]. The liver-determinant-extrahepatic (LDE) system applies to NAFLD and MAFLD and combines information on liver status independently of histology (L), determinants which include sex and reproductive status, genetic, and endocrine assessment (D), and extrahepatic manifestations at a metabolic, cardiovascular, and tumor level (E).
Table 3
Recently proposed sub-classifications of fatty liver disease (FLD) [84]
Acronym
Condition
Notes
MAFLD
Metabolic-associated fatty liver disease
Fatty liver because of the overarching metabolic syndrome. Can occur in lean individuals, in overweight/obese individuals, and in T2DM individuals [1012, 134]
O-MAFLD
Obesity-linked MAFLD
Subclassification of MAFLD. Defines the altered gluco-lipid metabolism and pro-inflammatory metabolic changes which link obesity to NAFLD [23, 40, 84, 135, 136]
SMAFLD
Sarcopenia-linked MAFLD
Subclassification of MAFLD. Fatty liver occurring during age-related, chronic progressive muscle mass loss, with or without obesity [137]. Sarcopenia can induce insulin resistance, steatosis and histological fibrosis grades [22]. Role for decreased skeletal muscle myokines involved in FFA metabolism [138]. Potential therapeutic target via the myostatin and/or activin/follistatin axis [139]. Can occur alone or combined with O-MAFLD, requiring double therapeutic approach
GAFLD
Genetics-associated fatty liver disease
Genetic variants either monogenic or polygenic not necessarily causing metabolic abnormalities [140]
LAFLD
Lipodystrophy-associated fatty liver disease
Fat or muscle mass abnormalities with partial or complete loss of adipose tissue and predisposition to develop fatty liver without obesity. Classified as congenital, familial partial or acquired disorders [84, 141]
AAFLD
Alcoholic-associated fatty liver disease
Occurs with significant chronic alcohol consumption. Similar potential progression as for NAFLD and risk of associated metabolic/cardiovascular complications [50, 84, 142]
CAFLD
Combined causes of FLD
Overlap of different entities of FLD, e.g., obesity and sarcopenia or genetic causes, or lipodystrophy
XAFLD
Yet-to-be-defined subgroups
Conditions of FLD where etiology remains (temporarily or constantly) undefined (e.g., autoimmune hepatitis [143]; viral hepatitis [144]), or plays an emerging role (e.g., thyroid subclinical dysfunction [145], mitochondrial dysfunction [40, 82, 146, 147]
As for MAFLD, however, any sub-classification linked to liver steatosis needs validation studies with terminology to be agreed upon [84]. While searching for better classification of liver steatosis, both researchers, clinicians, scientific societies, patients’ associations, and other stakeholders must be aware that change in terminology requires a better understanding of the molecular basis of the disease entity. Benefits of patients must be balanced along with novel risk stratification and characteristics of disease [87].

Conclusions and future perspectives

The rapid epidemiological increment and the global diffusion of NAFLD is a matter of major concern in terms of healthcare and social burden of disease. Both etiology and pathogenesis of NAFLD are largely unknown, and a multidisciplinary approach is required to handle a frequent liver disease still missing a definitive therapy, beside lifestyle, and maintenance or achievement of ideal body weight. The ongoing discussion urges to revise the terminology [11, 12] since the acronym NAFLD has several limitations: it suggests ignorance about true etiology, it remains an exclusion criterium, it can be ambiguous and misleading, and appears to stigmatize the affected individuals because of the word “alcoholic”. Since NAFLD is very often and closely associated with metabolic dysfunctions, the current view is to shift the acronym NAFLD to MAFLD, i.e., metabolic-associated fatty liver disease. MAFLD becomes an “active” diagnosis based on the presence of overweight/obesity or, in the lean subject, on the combination of metabolic dysfunctions which act as high‐risk factors for events. MAFLD is independent of alcohol intake and the co‐existing causes of liver disease. The change in terminology is likely to improve the classification of affected individuals, the disease awareness, the comprehension of terminology and pathophysiological aspects involved, the choice of more personalized therapeutic approaches, while avoiding the intrinsic stigmatization due to the term “non-alcoholic” (Table 4).
Table 4
Principal differences between NAFLD and MAFLD
Major features
NAFLD
MAFLD
Initial diagnosis of liver steatosis
Imaging
Histology
Imaging
Histology
Biomarkers
Terminology criteria
Based on “negative” assumptions
Based on “positive” assumptions
Terminology comprehension/disease awareness
Poor
Clear
Terminology stigma
Yes (“alcoholic”)
No
Alcohol intake criterium
Dependent
Not dependent
Metabolic dysfunction
Not required
Required
Combination with other liver diseases
No (excluded)
Possible, if present
Liver biopsy
Essential for diagnosis of NASH
Not required
Chance for multidisciplinary interactions
Medium
Maximum
Therapeutic reflections
Few
Several
Possibility to identify groups at increased risk
Moderate
Likely high for hepatic/extrahepatic complications (metabolic, cardiovascular, genetic risk)
Limited data in pediatric populations
Even more recently, other sub-classification have been proposed, e.g., the LDE terminology [128], and a detailed nomenclature has been proposed to concentrate the heterogeneous causes of fatty liver disease under one umbrella [84] (Table 3). We must take note of such shifts of paradigm and contribute to advance the discussion further. Several partners must agree upon a novel terminology, including clinicians, researchers, pharmaceutical industries, patients and their associations, and scientific societies. In this multidisciplinary field, we need motivated and dedicated researchers with holistic views, to bring tangible pathophysiological, diagnostic, and therapeutic benefits for the populations worldwide suffering from fatty liver disease and related burden of disease. Along with this shift of paradigm, i.e., NAFLD vs. MAFLD, the role of internal medicine and internists is undoubtedly gaining even more trust.

Acknowledgements

The author is indebted to Leonilde Bonfrate, Giuseppe Calamita, Agostino Di Ciaula, Domenica Di Palo, Dan Dumitrascu, Karel van Erpecum, Mohamad Khalil, Marcin Krawczyk, Paulo Oliveira, Harshita Shanmugam, and David Q.-H. Wang for helpful scientific discussion and longstanding collaborations.

Declarations

Conflict of interest

The author has no conflict of interest.

Human and animal rights statement and Informed consent

The article is a review analysis. Human Participants and/or Animals have not been involved in the present study.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Anästhesiologie

Kombi-Abonnement

Mit e.Med Anästhesiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes AINS, den Premium-Inhalten der AINS-Fachzeitschriften, inklusive einer gedruckten AINS-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Ludwig J, Viggiano TR, McGill DB, Oh BJ (1980) Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clinic proceedings. Mayo Clinic 55(7):434–438 Ludwig J, Viggiano TR, McGill DB, Oh BJ (1980) Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clinic proceedings. Mayo Clinic 55(7):434–438
2.
Zurück zum Zitat Schaffner F, Thaler H (1986) Nonalcoholic fatty liver disease. Prog Liver Dis 8:283–298PubMed Schaffner F, Thaler H (1986) Nonalcoholic fatty liver disease. Prog Liver Dis 8:283–298PubMed
9.
Zurück zum Zitat 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.29367CrossRefPubMed 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.​29367CrossRefPubMed
10.
Zurück zum Zitat Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wong VW-S, Dufour J-F, Schattenberg JM (2020) A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol 73(1):202–209CrossRefPubMed Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wong VW-S, Dufour J-F, Schattenberg JM (2020) A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol 73(1):202–209CrossRefPubMed
12.
Zurück zum Zitat Mendez-Sanchez N, Bugianesi E, Gish RG, Lammert F, Tilg H, Nguyen MH, Sarin SK, Fabrellas N, Zelber-Sagi S, Fan JG, Shiha G, Targher G, Zheng MH, Chan WK, Vinker S, Kawaguchi T, Castera L, Yilmaz Y, Korenjak M, Spearman CW, Ungan M, Palmer M, El-Shabrawi M, Gruss HJ, Dufour JF, Dhawan A, Wedemeyer H, George J, Valenti L, Fouad Y, Romero-Gomez M, Eslam M, Global multi-stakeholder consensus on the redefinition of fatty liver d (2022) Global multi-stakeholder endorsement of the MAFLD definition. Lancet Gastroenterol Hepatol 7(5):388–390. https://doi.org/10.1016/S2468-1253(22)00062-0CrossRefPubMed Mendez-Sanchez N, Bugianesi E, Gish RG, Lammert F, Tilg H, Nguyen MH, Sarin SK, Fabrellas N, Zelber-Sagi S, Fan JG, Shiha G, Targher G, Zheng MH, Chan WK, Vinker S, Kawaguchi T, Castera L, Yilmaz Y, Korenjak M, Spearman CW, Ungan M, Palmer M, El-Shabrawi M, Gruss HJ, Dufour JF, Dhawan A, Wedemeyer H, George J, Valenti L, Fouad Y, Romero-Gomez M, Eslam M, Global multi-stakeholder consensus on the redefinition of fatty liver d (2022) Global multi-stakeholder endorsement of the MAFLD definition. Lancet Gastroenterol Hepatol 7(5):388–390. https://​doi.​org/​10.​1016/​S2468-1253(22)00062-0CrossRefPubMed
13.
17.
Zurück zum Zitat Estes C, Anstee QM, Arias-Loste MT, Bantel H, Bellentani S, Caballeria J, Colombo M, Craxi A, Crespo J, Day CP, Eguchi Y, Geier A, Kondili LA, Kroy DC, Lazarus JV, Loomba R, Manns MP, Marchesini G, Nakajima A, Negro F, Petta S, Ratziu V, Romero-Gomez M, Sanyal A, Schattenberg JM, Tacke F, Tanaka J, Trautwein C, Wei L, Zeuzem S, Razavi H (2018) Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J Hepatol 69(4):896–904. https://doi.org/10.1016/j.jhep.2018.05.036CrossRefPubMed Estes C, Anstee QM, Arias-Loste MT, Bantel H, Bellentani S, Caballeria J, Colombo M, Craxi A, Crespo J, Day CP, Eguchi Y, Geier A, Kondili LA, Kroy DC, Lazarus JV, Loomba R, Manns MP, Marchesini G, Nakajima A, Negro F, Petta S, Ratziu V, Romero-Gomez M, Sanyal A, Schattenberg JM, Tacke F, Tanaka J, Trautwein C, Wei L, Zeuzem S, Razavi H (2018) Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J Hepatol 69(4):896–904. https://​doi.​org/​10.​1016/​j.​jhep.​2018.​05.​036CrossRefPubMed
23.
Zurück zum Zitat Polyzos SA, and Mantzoros CS (2020). Making progress in nonalcoholic fatty liver disease (NAFLD) as we are transitioning from the era of NAFLD to dys-metabolism associated fatty liver disease (DAFLD). Metab-Clin Exp 111 Polyzos SA, and Mantzoros CS (2020). Making progress in nonalcoholic fatty liver disease (NAFLD) as we are transitioning from the era of NAFLD to dys-metabolism associated fatty liver disease (DAFLD). Metab-Clin Exp 111
25.
30.
32.
Zurück zum Zitat Alferink LJM, Radjabzadeh D, Erler NS, Vojinovic D, Medina-Gomez C, Uitterlinden AG, de Knegt RJ, Amin N, Ikram MA, Janssen HLA, Kiefte-de Jong JC, Metselaar HJ, van Duijn CM, Kraaij R, Darwish Murad S (2021) Microbiomics, metabolomics, predicted metagenomics, and hepatic steatosis in a population-based study of 1,355 adults. Hepatology 73(3):968–982. https://doi.org/10.1002/hep.31417CrossRefPubMed Alferink LJM, Radjabzadeh D, Erler NS, Vojinovic D, Medina-Gomez C, Uitterlinden AG, de Knegt RJ, Amin N, Ikram MA, Janssen HLA, Kiefte-de Jong JC, Metselaar HJ, van Duijn CM, Kraaij R, Darwish Murad S (2021) Microbiomics, metabolomics, predicted metagenomics, and hepatic steatosis in a population-based study of 1,355 adults. Hepatology 73(3):968–982. https://​doi.​org/​10.​1002/​hep.​31417CrossRefPubMed
33.
Zurück zum Zitat Smyk W, Janik MK, Portincasa P, Milkiewicz P, Lammert F, Krawczyk M (2020) COVID-19: focus on the lungs but do not forget the gastrointestinal tract. Eur J Clin Investig 50(9):e13276CrossRef Smyk W, Janik MK, Portincasa P, Milkiewicz P, Lammert F, Krawczyk M (2020) COVID-19: focus on the lungs but do not forget the gastrointestinal tract. Eur J Clin Investig 50(9):e13276CrossRef
40.
Zurück zum Zitat Di Ciaula A, Passarella S, Shanmugam H, Noviello M, Bonfrate L, Wang DQ-H, Portincasa P (2021) Nonalcoholic fatty liver disease (NAFLD). Mitochondria as players and targets of therapies? Int J Mol Sci 22(10):5375CrossRefPubMedPubMedCentral Di Ciaula A, Passarella S, Shanmugam H, Noviello M, Bonfrate L, Wang DQ-H, Portincasa P (2021) Nonalcoholic fatty liver disease (NAFLD). Mitochondria as players and targets of therapies? Int J Mol Sci 22(10):5375CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Portincasa P, Wang DQH (2017) Nonalcoholic fatty liver and gallstone disease. In: Wang DQH, Portincasa P (eds) Gallstones. Recent advances in epidemiology, pathogenesis, diagnosis and management. Nova Science Publisher Inc, USA, pp 387–414 Portincasa P, Wang DQH (2017) Nonalcoholic fatty liver and gallstone disease. In: Wang DQH, Portincasa P (eds) Gallstones. Recent advances in epidemiology, pathogenesis, diagnosis and management. Nova Science Publisher Inc, USA, pp 387–414
43.
Zurück zum Zitat Grattagliano I, De Bari O, Di Palo D, Montecucco F, Carbone F, Oliveira P, Wang DQH, Portincasa P (2018) Mitochondria in liver diseases. In: Oliveira P (ed) Mitochondrial biology and experimental therapeutics. Springer Nature, Berlin, pp 91–126CrossRef Grattagliano I, De Bari O, Di Palo D, Montecucco F, Carbone F, Oliveira P, Wang DQH, Portincasa P (2018) Mitochondria in liver diseases. In: Oliveira P (ed) Mitochondrial biology and experimental therapeutics. Springer Nature, Berlin, pp 91–126CrossRef
45.
Zurück zum Zitat Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, Sebastiani G, Ekstedt M, Hagstrom H, Nasr P, Stal P, Wong VW, Kechagias S, Hultcrantz R, Loomba R (2017) Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology 65(5):1557–1565. https://doi.org/10.1002/hep.29085CrossRefPubMed Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, Sebastiani G, Ekstedt M, Hagstrom H, Nasr P, Stal P, Wong VW, Kechagias S, Hultcrantz R, Loomba R (2017) Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology 65(5):1557–1565. https://​doi.​org/​10.​1002/​hep.​29085CrossRefPubMed
47.
Zurück zum Zitat Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ, American Gastroenterological A, American Association for the Study of Liver D, and American College of G (2012) The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American gastroenterological association, American association for the study of liver diseases, and American college of gastroenterology. Gastroenterology 142(7):1592–1609. https://doi.org/10.1053/j.gastro.2012.04.001CrossRefPubMed Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ, American Gastroenterological A, American Association for the Study of Liver D, and American College of G (2012) The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American gastroenterological association, American association for the study of liver diseases, and American college of gastroenterology. Gastroenterology 142(7):1592–1609. https://​doi.​org/​10.​1053/​j.​gastro.​2012.​04.​001CrossRefPubMed
48.
Zurück zum Zitat Blomdahl J, Nasr P, Ekstedt M, Kechagias S (2021) Moderate alcohol consumption is associated with advanced fibrosis in non-alcoholic fatty liver disease and shows a synergistic effect with type 2 diabetes mellitus. Metab Clin Exp 115:154439CrossRefPubMed Blomdahl J, Nasr P, Ekstedt M, Kechagias S (2021) Moderate alcohol consumption is associated with advanced fibrosis in non-alcoholic fatty liver disease and shows a synergistic effect with type 2 diabetes mellitus. Metab Clin Exp 115:154439CrossRefPubMed
51.
52.
62.
Zurück zum Zitat Yarnoz-Esquiroz P, Olazaran L, Aguas-Ayesa M, Perdomo CM, Garcia-Goni M, Silva C, Fernandez-Formoso JA, Escalada J, Montecucco F, Portincasa P, Fruhbeck G (2022) “Obesities”: position statement on a complex disease entity with multifaceted drivers. Eur J Clin Investig 52(7):e13811. https://doi.org/10.1111/eci.13811CrossRef Yarnoz-Esquiroz P, Olazaran L, Aguas-Ayesa M, Perdomo CM, Garcia-Goni M, Silva C, Fernandez-Formoso JA, Escalada J, Montecucco F, Portincasa P, Fruhbeck G (2022) “Obesities”: position statement on a complex disease entity with multifaceted drivers. Eur J Clin Investig 52(7):e13811. https://​doi.​org/​10.​1111/​eci.​13811CrossRef
70.
Zurück zum Zitat Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, Marczak L, Mokdad AH, Moradi-Lakeh M, Naghavi M, Salama JS, Vos T, Abate KH, Abbafati C, Ahmed MB, Al-Aly Z, Alkerwi A, Al-Raddadi R, Amare AT, Amberbir A, Amegah AK, Amini E, Amrock SM, Anjana RM, Arnlov J, Asayesh H, Banerjee A, Barac A, Baye E, Bennett DA, Beyene AS, Biadgilign S, Biryukov S, Bjertness E, Boneya DJ, Campos-Nonato I, Carrero JJ, Cecilio P, Cercy K, Ciobanu LG, Cornaby L, Damtew SA, Dandona L, Dandona R, Dharmaratne SD, Duncan BB, Eshrati B, Esteghamati A, Feigin VL, Fernandes JC, Furst T, Gebrehiwot TT, Gold A, Gona PN, Goto A, Habtewold TD, Hadush KT, Hafezi-Nejad N, Hay SI, Horino M, Islami F, Kamal R, Kasaeian A, Katikireddi SV, Kengne AP, Kesavachandran CN, Khader YS, Khang YH, Khubchandani J, Kim D, Kim YJ, Kinfu Y, Kosen S, Ku T, Defo BK, Kumar GA, Larson HJ, Leinsalu M, Liang X, Lim SS, Liu P, Lopez AD, Lozano R, Majeed A, Malekzadeh R, Malta DC, Mazidi M, McAlinden C, McGarvey ST, Mengistu DT, Mensah GA, Mensink GBM, Mezgebe HB, Mirrakhimov EM, Mueller UO, Noubiap JJ, Obermeyer CM, Ogbo FA, Owolabi MO, Patton GC, Pourmalek F, Qorbani M, Rafay A, Rai RK, Ranabhat CL, Reinig N, Safiri S, Salomon JA, Sanabria JR, Santos IS, Sartorius B, Sawhney M, Schmidhuber J, Schutte AE, Schmidt MI, Sepanlou SG, Shamsizadeh M, Sheikhbahaei S, Shin MJ, Shiri R, Shiue I, Roba HS, Silva DAS, Silverberg JI, Singh JA, Stranges S, Swaminathan S, Tabares-Seisdedos R, Tadese F, Tedla BA, Tegegne BS, Terkawi AS, Thakur JS, Tonelli M, Topor-Madry R, Tyrovolas S, Ukwaja KN, Uthman OA, Vaezghasemi M, Vasankari T, Vlassov VV, Vollset SE, Weiderpass E, Werdecker A, Wesana J, Westerman R, Yano Y, Yonemoto N, Yonga G, Zaidi Z, Zenebe ZM, Zipkin B, Murray CJL, GBD 215 Obesity Collaborators (2017) Health effects of overweight and obesity in 195 Countries over 25 years. New Engl J Med 377(1):13–27. https://doi.org/10.1056/NEJMoa1614362CrossRefPubMed Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, Marczak L, Mokdad AH, Moradi-Lakeh M, Naghavi M, Salama JS, Vos T, Abate KH, Abbafati C, Ahmed MB, Al-Aly Z, Alkerwi A, Al-Raddadi R, Amare AT, Amberbir A, Amegah AK, Amini E, Amrock SM, Anjana RM, Arnlov J, Asayesh H, Banerjee A, Barac A, Baye E, Bennett DA, Beyene AS, Biadgilign S, Biryukov S, Bjertness E, Boneya DJ, Campos-Nonato I, Carrero JJ, Cecilio P, Cercy K, Ciobanu LG, Cornaby L, Damtew SA, Dandona L, Dandona R, Dharmaratne SD, Duncan BB, Eshrati B, Esteghamati A, Feigin VL, Fernandes JC, Furst T, Gebrehiwot TT, Gold A, Gona PN, Goto A, Habtewold TD, Hadush KT, Hafezi-Nejad N, Hay SI, Horino M, Islami F, Kamal R, Kasaeian A, Katikireddi SV, Kengne AP, Kesavachandran CN, Khader YS, Khang YH, Khubchandani J, Kim D, Kim YJ, Kinfu Y, Kosen S, Ku T, Defo BK, Kumar GA, Larson HJ, Leinsalu M, Liang X, Lim SS, Liu P, Lopez AD, Lozano R, Majeed A, Malekzadeh R, Malta DC, Mazidi M, McAlinden C, McGarvey ST, Mengistu DT, Mensah GA, Mensink GBM, Mezgebe HB, Mirrakhimov EM, Mueller UO, Noubiap JJ, Obermeyer CM, Ogbo FA, Owolabi MO, Patton GC, Pourmalek F, Qorbani M, Rafay A, Rai RK, Ranabhat CL, Reinig N, Safiri S, Salomon JA, Sanabria JR, Santos IS, Sartorius B, Sawhney M, Schmidhuber J, Schutte AE, Schmidt MI, Sepanlou SG, Shamsizadeh M, Sheikhbahaei S, Shin MJ, Shiri R, Shiue I, Roba HS, Silva DAS, Silverberg JI, Singh JA, Stranges S, Swaminathan S, Tabares-Seisdedos R, Tadese F, Tedla BA, Tegegne BS, Terkawi AS, Thakur JS, Tonelli M, Topor-Madry R, Tyrovolas S, Ukwaja KN, Uthman OA, Vaezghasemi M, Vasankari T, Vlassov VV, Vollset SE, Weiderpass E, Werdecker A, Wesana J, Westerman R, Yano Y, Yonemoto N, Yonga G, Zaidi Z, Zenebe ZM, Zipkin B, Murray CJL, GBD 215 Obesity Collaborators (2017) Health effects of overweight and obesity in 195 Countries over 25 years. New Engl J Med 377(1):13–27. https://​doi.​org/​10.​1056/​NEJMoa1614362CrossRefPubMed
83.
Zurück zum Zitat Shiha G, Korenjak M, Eskridge W, Casanovas T, Velez-Moller P, Hogstrom S, Richardson B, Munoz C, Sigurethardottir S, Coulibaly A, Milan M, Bautista F, Leung NWY, Mooney V, Obekpa S, Bech E, Polavarapu N, Hamed AE, Radiani T, Purwanto E, Bright B, Ali M, Dovia CK, McColaugh L, Koulla Y, Dufour JF, Soliman R, Eslam M (2021) Redefining fatty liver disease: an international patient perspective. Lancet Gastroenterol Hepatol 6(1):73–79. https://doi.org/10.1016/S2468-1253(20)30294-6CrossRefPubMed Shiha G, Korenjak M, Eskridge W, Casanovas T, Velez-Moller P, Hogstrom S, Richardson B, Munoz C, Sigurethardottir S, Coulibaly A, Milan M, Bautista F, Leung NWY, Mooney V, Obekpa S, Bech E, Polavarapu N, Hamed AE, Radiani T, Purwanto E, Bright B, Ali M, Dovia CK, McColaugh L, Koulla Y, Dufour JF, Soliman R, Eslam M (2021) Redefining fatty liver disease: an international patient perspective. Lancet Gastroenterol Hepatol 6(1):73–79. https://​doi.​org/​10.​1016/​S2468-1253(20)30294-6CrossRefPubMed
85.
Zurück zum Zitat Alqahtani SA, Paik JM, Biswas R, Arshad T, Henry L, Younossi ZM (2021) Poor awareness of liver disease among adults with NAFLD in the United States. Hepatol Commun 5(11):1833–1847CrossRefPubMedPubMedCentral Alqahtani SA, Paik JM, Biswas R, Arshad T, Henry L, Younossi ZM (2021) Poor awareness of liver disease among adults with NAFLD in the United States. Hepatol Commun 5(11):1833–1847CrossRefPubMedPubMedCentral
89.
Zurück zum Zitat Nan Y, An J, Bao J, Chen H, Chen Y, Ding H, Dou X, Duan Z, Fan J, Gao Y, Han T, Han Y, Hu P, Huang Y, Huang Y, Jia J, Jiang J, Jiang Y, Li J, Li J, Li R, Li S, Li W, Li Y, Lin S, Liu J, Liu S, Lu L, Lu Q, Luo X, Ma X, Rao H, Ren H, Ren W, Shang J, Shi L, Su M, Wang B, Wang R, Wei L, Wen Z, Wu B, Wu J, Xin S, Xing H, Xu J, Yan M, Yang J, Yang J, Yang L, Yang Y, Yu Y, Zhang L, Zhang L, Zhang X, Zhang Y, Zhang Y, Zhao J, Zhao S, Zheng H, Zhou Y, Zhou Y, Zhuang H, Zuo W, Xu X, Qiao L (2021) The Chinese Society of Hepatology position statement on the redefinition of fatty liver disease. J Hepatol 75(2):454–461. https://doi.org/10.1016/j.jhep.2021.05.003CrossRefPubMed Nan Y, An J, Bao J, Chen H, Chen Y, Ding H, Dou X, Duan Z, Fan J, Gao Y, Han T, Han Y, Hu P, Huang Y, Huang Y, Jia J, Jiang J, Jiang Y, Li J, Li J, Li R, Li S, Li W, Li Y, Lin S, Liu J, Liu S, Lu L, Lu Q, Luo X, Ma X, Rao H, Ren H, Ren W, Shang J, Shi L, Su M, Wang B, Wang R, Wei L, Wen Z, Wu B, Wu J, Xin S, Xing H, Xu J, Yan M, Yang J, Yang J, Yang L, Yang Y, Yu Y, Zhang L, Zhang L, Zhang X, Zhang Y, Zhang Y, Zhao J, Zhao S, Zheng H, Zhou Y, Zhou Y, Zhuang H, Zuo W, Xu X, Qiao L (2021) The Chinese Society of Hepatology position statement on the redefinition of fatty liver disease. J Hepatol 75(2):454–461. https://​doi.​org/​10.​1016/​j.​jhep.​2021.​05.​003CrossRefPubMed
90.
Zurück zum Zitat Eslam M, Sarin SK, Wong VW, Fan JG, Kawaguchi T, Ahn SH, Zheng MH, Shiha G, Yilmaz Y, Gani R, Alam S, Dan YY, Kao JH, Hamid S, Cua IH, Chan WK, Payawal D, Tan SS, Tanwandee T, Adams LA, Kumar M, Omata M, George J (2020) The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatol Int 14(6):889–919. https://doi.org/10.1007/s12072-020-10094-2CrossRefPubMed Eslam M, Sarin SK, Wong VW, Fan JG, Kawaguchi T, Ahn SH, Zheng MH, Shiha G, Yilmaz Y, Gani R, Alam S, Dan YY, Kao JH, Hamid S, Cua IH, Chan WK, Payawal D, Tan SS, Tanwandee T, Adams LA, Kumar M, Omata M, George J (2020) The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatol Int 14(6):889–919. https://​doi.​org/​10.​1007/​s12072-020-10094-2CrossRefPubMed
91.
Zurück zum Zitat Shiha G, Alswat K, Al Khatry M, Sharara AI, Ormeci N, Waked I, Benazzouz M, Al-Ali F, Hamed AE, Hamoudi W, Attia D, Derbala M, Sharaf-Eldin M, Al-Busafi SA, Zaky S, Bamakhrama K, Ibrahim N, Ajlouni Y, Sabbah M, Salama M, Anushiravani A, Afredj N, Barakat S, Hashim A, Fouad Y, Soliman R (2021) Nomenclature and definition of metabolic-associated fatty liver disease: a consensus from the Middle East and north Africa. Lancet Gastroenterol Hepatol 6(1):57–64. https://doi.org/10.1016/S2468-1253(20)30213-2CrossRefPubMed Shiha G, Alswat K, Al Khatry M, Sharara AI, Ormeci N, Waked I, Benazzouz M, Al-Ali F, Hamed AE, Hamoudi W, Attia D, Derbala M, Sharaf-Eldin M, Al-Busafi SA, Zaky S, Bamakhrama K, Ibrahim N, Ajlouni Y, Sabbah M, Salama M, Anushiravani A, Afredj N, Barakat S, Hashim A, Fouad Y, Soliman R (2021) Nomenclature and definition of metabolic-associated fatty liver disease: a consensus from the Middle East and north Africa. Lancet Gastroenterol Hepatol 6(1):57–64. https://​doi.​org/​10.​1016/​S2468-1253(20)30213-2CrossRefPubMed
97.
Zurück zum Zitat Park H, Yoon EL, Cho S, Jun DW, Nah E-H (2022) Diabetes is the strongest risk factor of hepatic fibrosis in lean patients with non-alcoholic fatty liver disease. Gut 71(5):1035–1036CrossRefPubMed Park H, Yoon EL, Cho S, Jun DW, Nah E-H (2022) Diabetes is the strongest risk factor of hepatic fibrosis in lean patients with non-alcoholic fatty liver disease. Gut 71(5):1035–1036CrossRefPubMed
98.
Zurück zum Zitat Kawaguchi T, Torimura T (2020) Is metabolic syndrome responsible for the progression from NAFLD to NASH in non-obese patients? SpringerCrossRef Kawaguchi T, Torimura T (2020) Is metabolic syndrome responsible for the progression from NAFLD to NASH in non-obese patients? SpringerCrossRef
99.
Zurück zum Zitat Ochiai Y, Kawamura Y, Kobayashi M, Shindoh J, Kobayashi Y, Okubo S, Muraishi N, Kajiwara A, Iritani S, Fujiyama S, Hosaka T, Saitoh S, Sezaki H, Akuta N, Suzuki F, Suzuki Y, Ikeda K, Arase Y, Hashimoto M, Kumada H (2021) Effects of alcohol consumption on multiple hepatocarcinogenesis in patients with fatty liver disease. Hepatol Res 51(1):62–68. https://doi.org/10.1111/hepr.13572CrossRefPubMed Ochiai Y, Kawamura Y, Kobayashi M, Shindoh J, Kobayashi Y, Okubo S, Muraishi N, Kajiwara A, Iritani S, Fujiyama S, Hosaka T, Saitoh S, Sezaki H, Akuta N, Suzuki F, Suzuki Y, Ikeda K, Arase Y, Hashimoto M, Kumada H (2021) Effects of alcohol consumption on multiple hepatocarcinogenesis in patients with fatty liver disease. Hepatol Res 51(1):62–68. https://​doi.​org/​10.​1111/​hepr.​13572CrossRefPubMed
100.
Zurück zum Zitat Xu L, Xie J, Chen S, Chen Y, Yang H, Miao M, Zhu Z, Li Y, Yu C, Xu C (2020) Light-to-moderate alcohol consumption is associated with increased risk of type 2 diabetes in individuals with nonalcoholic fatty liver disease: a nine-year cohort study. Off J Am College Gastroenterol ACG 115(6):876–884CrossRef Xu L, Xie J, Chen S, Chen Y, Yang H, Miao M, Zhu Z, Li Y, Yu C, Xu C (2020) Light-to-moderate alcohol consumption is associated with increased risk of type 2 diabetes in individuals with nonalcoholic fatty liver disease: a nine-year cohort study. Off J Am College Gastroenterol ACG 115(6):876–884CrossRef
103.
Zurück zum Zitat Fuster D, Samet JH (2018) Alcohol use in patients with chronic liver disease. New Engl J Med 379(13):1251–1261CrossRefPubMed Fuster D, Samet JH (2018) Alcohol use in patients with chronic liver disease. New Engl J Med 379(13):1251–1261CrossRefPubMed
104.
Zurück zum Zitat Choi HS, Brouwer WP, Zanjir WM, de Man RA, Feld JJ, Hansen BE, Janssen HL, Patel K (2020) Nonalcoholic steatohepatitis is associated with liver-related outcomes and all-cause mortality in chronic hepatitis B. Hepatology 71(2):539–548CrossRefPubMed Choi HS, Brouwer WP, Zanjir WM, de Man RA, Feld JJ, Hansen BE, Janssen HL, Patel K (2020) Nonalcoholic steatohepatitis is associated with liver-related outcomes and all-cause mortality in chronic hepatitis B. Hepatology 71(2):539–548CrossRefPubMed
105.
Zurück zum Zitat Tada T, Nishimura T, Matono T, Yoshida M, Yuri M, Fujiwara A, Yuri Y, Takashima T, Aizawa N, Ikeda N, Enomoto H, Kumada T, Iijima H (2021) Association of liver stiffness and steatosis with hepatocellular carcinoma development in patients with hepatitis C virus infection who received direct-acting antiviral therapy and achieved sustained virological response. Hepatol Res 51(8):860–869. https://doi.org/10.1111/hepr.13677CrossRefPubMed Tada T, Nishimura T, Matono T, Yoshida M, Yuri M, Fujiwara A, Yuri Y, Takashima T, Aizawa N, Ikeda N, Enomoto H, Kumada T, Iijima H (2021) Association of liver stiffness and steatosis with hepatocellular carcinoma development in patients with hepatitis C virus infection who received direct-acting antiviral therapy and achieved sustained virological response. Hepatol Res 51(8):860–869. https://​doi.​org/​10.​1111/​hepr.​13677CrossRefPubMed
106.
Zurück zum Zitat Guerreiro GTS, Longo L, Fonseca MA, de Souza VEG, Alvares-da-Silva MR (2021) Does the risk of cardiovascular events differ between biopsy-proven NAFLD and MAFLD? Hepatol Int 15(2):380–391CrossRefPubMed Guerreiro GTS, Longo L, Fonseca MA, de Souza VEG, Alvares-da-Silva MR (2021) Does the risk of cardiovascular events differ between biopsy-proven NAFLD and MAFLD? Hepatol Int 15(2):380–391CrossRefPubMed
108.
Zurück zum Zitat Pavlik L, Regev A, Ardayfio PA, Chalasani NP (2019) Drug-induced steatosis and steatohepatitis: the search for novel serum biomarkers among potential biomarkers for non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Drug Saf 42(6):701–711CrossRefPubMed Pavlik L, Regev A, Ardayfio PA, Chalasani NP (2019) Drug-induced steatosis and steatohepatitis: the search for novel serum biomarkers among potential biomarkers for non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Drug Saf 42(6):701–711CrossRefPubMed
115.
Zurück zum Zitat Tsutsumi T, Eslam M, Kawaguchi T, Yamamura S, Kawaguchi A, Nakano D, Koseki M, Yoshinaga S, Takahashi H, Anzai K, George J, Torimura T (2021) MAFLD better predicts the progression of atherosclerotic cardiovascular risk than NAFLD: generalized estimating equation approach. Hepatol Res 51(11):1115–1128. https://doi.org/10.1111/hepr.13685CrossRefPubMed Tsutsumi T, Eslam M, Kawaguchi T, Yamamura S, Kawaguchi A, Nakano D, Koseki M, Yoshinaga S, Takahashi H, Anzai K, George J, Torimura T (2021) MAFLD better predicts the progression of atherosclerotic cardiovascular risk than NAFLD: generalized estimating equation approach. Hepatol Res 51(11):1115–1128. https://​doi.​org/​10.​1111/​hepr.​13685CrossRefPubMed
116.
Zurück zum Zitat Eslam M, Ahmed A, Despres JP, Jha V, Halford JCG, Wei Chieh JT, Harris DCH, Nangaku M, Colagiuri S, Targher G, Joshi S, Byrne CD, Khunti K, Nguyen MH, Gish RG, George J (2021) Incorporating fatty liver disease in multidisciplinary care and novel clinical trial designs for patients with metabolic diseases. Lancet Gastroenterol Hepatol 6(9):743–753. https://doi.org/10.1016/S2468-1253(21)00132-1CrossRefPubMed Eslam M, Ahmed A, Despres JP, Jha V, Halford JCG, Wei Chieh JT, Harris DCH, Nangaku M, Colagiuri S, Targher G, Joshi S, Byrne CD, Khunti K, Nguyen MH, Gish RG, George J (2021) Incorporating fatty liver disease in multidisciplinary care and novel clinical trial designs for patients with metabolic diseases. Lancet Gastroenterol Hepatol 6(9):743–753. https://​doi.​org/​10.​1016/​S2468-1253(21)00132-1CrossRefPubMed
119.
Zurück zum Zitat Mendez-Sanchez N, Arrese M, Gadano A, Oliveira CP, Fassio E, Arab JP, Chavez-Tapia NC, Dirchwolf M, Torre A, Ridruejo E, Pinchemel-Cotrim H, Castellanos Fernandez MI, Uribe M, Girala M, Diaz-Ferrer J, Restrepo JC, Padilla-Machaca M, Dagher L, Gatica M, Olaechea B, Pessoa MG, Silva M (2021) The Latin American association for the study of the liver (ALEH) position statement on the redefinition of fatty liver disease. Lancet Gastroenterol Hepatol 6(1):65–72. https://doi.org/10.1016/S2468-1253(20)30340-XCrossRefPubMed Mendez-Sanchez N, Arrese M, Gadano A, Oliveira CP, Fassio E, Arab JP, Chavez-Tapia NC, Dirchwolf M, Torre A, Ridruejo E, Pinchemel-Cotrim H, Castellanos Fernandez MI, Uribe M, Girala M, Diaz-Ferrer J, Restrepo JC, Padilla-Machaca M, Dagher L, Gatica M, Olaechea B, Pessoa MG, Silva M (2021) The Latin American association for the study of the liver (ALEH) position statement on the redefinition of fatty liver disease. Lancet Gastroenterol Hepatol 6(1):65–72. https://​doi.​org/​10.​1016/​S2468-1253(20)30340-XCrossRefPubMed
120.
Zurück zum Zitat Eslam M, Alkhouri N, Vajro P, Baumann U, Weiss R, Socha P, Marcus C, Lee WS, Kelly D, Porta G, El-Guindi MA, Alisi A, Mann JP, Mouane N, Baur LA, Dhawan A, George J (2021) Defining paediatric metabolic (dysfunction)-associated fatty liver disease: an international expert consensus statement. Lancet Gastroenterol Hepatol 6(10):864–873. https://doi.org/10.1016/S2468-1253(21)00183-7CrossRefPubMed Eslam M, Alkhouri N, Vajro P, Baumann U, Weiss R, Socha P, Marcus C, Lee WS, Kelly D, Porta G, El-Guindi MA, Alisi A, Mann JP, Mouane N, Baur LA, Dhawan A, George J (2021) Defining paediatric metabolic (dysfunction)-associated fatty liver disease: an international expert consensus statement. Lancet Gastroenterol Hepatol 6(10):864–873. https://​doi.​org/​10.​1016/​S2468-1253(21)00183-7CrossRefPubMed
124.
Zurück zum Zitat Karlsen TH, Sheron N, Zelber-Sagi S, Carrieri P, Dusheiko G, Bugianesi E, Pryke R, Hutchinson SJ, Sangro B, Martin NK, Cecchini M, Dirac MA, Belloni A, Serra-Burriel M, Ponsioen CY, Sheena B, Lerouge A, Devaux M, Scott N, Hellard M, Verkade HJ, Sturm E, Marchesini G, Yki-Jarvinen H, Byrne CD, Targher G, Tur-Sinai A, Barrett D, Ninburg M, Reic T, Taylor A, Rhodes T, Treloar C, Petersen C, Schramm C, Flisiak R, Simonova MY, Pares A, Johnson P, Cucchetti A, Graupera I, Lionis C, Pose E, Fabrellas N, Ma AT, Mendive JM, Mazzaferro V, Rutter H, Cortez-Pinto H, Kelly D, Burton R, Lazarus JV, Gines P, Buti M, Newsome PN, Burra P, Manns MP (2022) The EASL-lancet liver commission: protecting the next generation of Europeans against liver disease complications and premature mortality. Lancet 399(10319):61–116. https://doi.org/10.1016/S0140-6736(21)01701-3CrossRefPubMed Karlsen TH, Sheron N, Zelber-Sagi S, Carrieri P, Dusheiko G, Bugianesi E, Pryke R, Hutchinson SJ, Sangro B, Martin NK, Cecchini M, Dirac MA, Belloni A, Serra-Burriel M, Ponsioen CY, Sheena B, Lerouge A, Devaux M, Scott N, Hellard M, Verkade HJ, Sturm E, Marchesini G, Yki-Jarvinen H, Byrne CD, Targher G, Tur-Sinai A, Barrett D, Ninburg M, Reic T, Taylor A, Rhodes T, Treloar C, Petersen C, Schramm C, Flisiak R, Simonova MY, Pares A, Johnson P, Cucchetti A, Graupera I, Lionis C, Pose E, Fabrellas N, Ma AT, Mendive JM, Mazzaferro V, Rutter H, Cortez-Pinto H, Kelly D, Burton R, Lazarus JV, Gines P, Buti M, Newsome PN, Burra P, Manns MP (2022) The EASL-lancet liver commission: protecting the next generation of Europeans against liver disease complications and premature mortality. Lancet 399(10319):61–116. https://​doi.​org/​10.​1016/​S0140-6736(21)01701-3CrossRefPubMed
126.
Zurück zum Zitat Chan KE, Koh TJL, Tang ASP, Quek J, Yong JN, Tay P, Tan DJH, Lim WH, Lin SY, Huang D, Chan M, Khoo CM, Chew NWS, Kaewdech A, Chamroonkul N, Dan YY, Noureddin M, Muthiah M, Eslam M, Ng CH (2022) Global prevalence and clinical characteristics of metabolic-associated fatty liver disease: a meta-analysis and systematic review of 10 739 607 individuals. J Clin EndocrinolMetab 107(9):2691–2700. https://doi.org/10.1210/clinem/dgac321CrossRef Chan KE, Koh TJL, Tang ASP, Quek J, Yong JN, Tay P, Tan DJH, Lim WH, Lin SY, Huang D, Chan M, Khoo CM, Chew NWS, Kaewdech A, Chamroonkul N, Dan YY, Noureddin M, Muthiah M, Eslam M, Ng CH (2022) Global prevalence and clinical characteristics of metabolic-associated fatty liver disease: a meta-analysis and systematic review of 10 739 607 individuals. J Clin EndocrinolMetab 107(9):2691–2700. https://​doi.​org/​10.​1210/​clinem/​dgac321CrossRef
127.
Metadaten
Titel
NAFLD, MAFLD, and beyond: one or several acronyms for better comprehension and patient care
verfasst von
Piero Portincasa
Publikationsdatum
17.02.2023
Verlag
Springer International Publishing
Erschienen in
Internal and Emergency Medicine / Ausgabe 4/2023
Print ISSN: 1828-0447
Elektronische ISSN: 1970-9366
DOI
https://doi.org/10.1007/s11739-023-03203-0

Weitere Artikel der Ausgabe 4/2023

Internal and Emergency Medicine 4/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.