Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jun 27, 2022; 14(6): 1087-1098
Published online Jun 27, 2022. doi: 10.4254/wjh.v14.i6.1087
Metabolic-associated fatty liver disease from childhood to adulthood: State of art and future directions
Francesca Lanzaro, Stefano Guarino, Elisabetta D'Addio, Alessandra Salvatori, Josè Alberto D'Anna, Pierluigi Marzuillo, Emanuele Miraglia del Giudice, Anna Di Sessa, Department of Woman, Child, and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Naples 80138, Italy
ORCID number: Francesca Lanzaro (0000-0002-0976-6833); Stefano Guarino (0000-0002-0551-5236); Elisabetta D'addio (0000-002-2325-5255); Alessandra Salvatori (0000-0002-6289-2249); Josè Alberto D'anna (0000-0002-9922-0163); Pierluigi Marzuillo (0000-0003-4682-0170); Emanuele Miraglia del Giudice (0000-0002-1492-076X); Anna Di Sessa (0000-0002-5877-3757).
Author contributions: Lanzaro F wrote the first draft of the manuscript; Miraglia del Giudice E, Di Sessa A, and Marzuillo P conceived the manuscript; Di Sessa A, Miraglia del Giudice E, and Marzuillo P supervised the manuscript drafting; D'Addio E, Salvatori A, Guarino S, and D'Anna JA reviewed the literature data; Guarino S and Lanzaro F prepared the tables; all author contributed important intellectual content during manuscript drafting or revision.
Conflict-of-interest statement: There are no conflicts of interest to report.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Pierluigi Marzuillo, MD, PhD, Assistant Professor, Department of Woman, Child, and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via de Crecchio, 2, Naples 80138, Italy. pierluigi.marzuillo@gmail.com
Received: October 18, 2021
Peer-review started: October 18, 2021
First decision: December 3, 2021
Revised: December 26, 2021
Accepted: April 25, 2022
Article in press: April 25, 2022
Published online: June 27, 2022

Abstract

In 2020, an international group of experts proposed to replace the term of nonalcoholic fatty liver disease with metabolic-associated fatty liver disease (MAFLD). This recent proposal reflects the close association of fatty liver with metabolic derangements, as demonstrated by previous robust data. Several factors [including genetics, inflammation, metabolic abnormalities, insulin resistance (IR), obesity, prenatal determinants, and gut–liver axis] have been found to be involved in MAFLD pathophysiology, but this tangled puzzle remains to be clearly understood. In particular, IR has been recognized as a key player in metabolic impairments development in children with fatty liver. On this ground, MAFLD definition focuses on the pathophysiological basis of the disease, by emphasizing the crucial role of metabolic impairments in this condition. Although primarily developed for adults, MAFLD diagnostic criteria have been recently updated with an age-appropriate definition for sex and age percentiles, because of the increasing attention to cardiometabolic risk in childhood. To date, accumulating evidence is available on the feasibility of MAFLD definition in clinical practice, but some data are still conflicting in highly selected populations. Considering the growing prevalence worldwide of fatty liver and its close relationship with metabolic dysfunction both in children and adults with subsequent increased cardiovascular risk, early strategies for MAFLD identification, treatment and prevention are needed. Novel therapeutic insights for MAFLD based on promising innovative biological techniques are also emerging. We aimed to summarize the most recent evidence in this intriguing research area both in children and adults.

Key Words: Metabolic, dysfunction, Fatty, Liver, Pathophysiology, Cardiovascular, Risk, Adults, Children

Core tip: Recently, experts have proposed to rename nonalcoholic fatty liver disease as metabolic-associated fatty liver disease (MAFLD), by emphasizing the close association of fatty liver with the metabolic milieu. Given that, a growing number of studies have tested the effectiveness of the new definition in adults and children, although evidence in this latter population is still limited. However, expanding knowledge about MAFLD and its pathophysiology is crucial for a better identification of subjects at greater metabolic risk.



INTRODUCTION

As proposed by an international consensus in 2020[1], the nomenclature of nonalcoholic fatty liver disease (NAFLD) has been updated to metabolic-associated fatty liver disease (MAFLD). MAFLD diagnosis is based on histological (biopsy), imaging or blood biomarker evidence of hepatic steatosis, and on the presence of any condition among: (1) Overweight/obesity; (2) diabetes mellitus; or (3) evidence of metabolic dysregulation[1], commonly defined as ≥ 2 of these characteristics: (1) Waist circumference ≥ 102 cm in Caucasian male subjects and 88 cm in women (or ≥ 90/80 cm in Asian individuals); (2) blood pressure ≥ 130/85 mmHg or specific drug treatment; (3) triglyceride level ≥ 1.70 mmol/L or specific drug treatment; (4) high-density lipoprotein (HDL)-cholesterol < 1.0 mmol/L for men and < 1.3 mmol/L for women; (5) prediabetes (i.e., fasting glucose levels 5.6–6.9 mmol/L, or 2-h post-load glucose levels 7.8–11.0 mmol/L or hemoglobin A1c 5.7%–6.4%; (6) homeostasis model assessment-insulin resistance (HOMA-IR) score ≥ 2.5; and (7) high-sensitive C-reactive protein (hs-CRP) level > 2 mg/L.

Numerous different factors such as inflammation, sex, age, ethnicity, diet, microbiota, hormones, and genetics have been pathogenically linked to NAFLD[2-4], but current knowledge about MAFLD pathophysiology is still limited[5-6].

During the past decades, research focused on the strong association between insulin resistance (IR) and NAFLD[7]. In particular, previous data have largely supported the role of NAFLD as a hepatic manifestation of systemic metabolic disorders[2,3]. Based on these premises, the new nomenclature aims to strengthened the close association of fatty liver with metabolic dysfunction[2,8-12] to identify early subjects at higher risk of long-term metabolic consequences.

As noted for obesity and its related consequences [e.g., metabolic syndrome (MetS) and Type 2 diabetes (T2D)[13-15]], a key pathogenic role has been described for the low-grade systemic inflammation in modulating fibrosis development and the overall course of the hepatic disease. As a result, an inflammatory biomarker such as hs-CRP, has been considered as a MAFLD diagnostic criterion. However, it should be kept in mind that further specific diagnostic criteria for MetS define this peculiar cluster of metabolic abnormalities, according to age group[16,18]. In fact, the MetS definition provided for adults and children aged ≥ 10 years by the International Diabetes Federation (IDF)[16,17] was further integrated for subjects aged 2–11 years (Table 1). The comparison between MetS and MAFLD criteria (Tables 2 and 3) allows identification of MetS subjects with fatty liver as MAFLD patients. Although both conditions allow identification of subjects at higher cardiometabolic risk, the inclusion of fatty liver as a MAFLD criterion enhances the multifactorial pathophysiology of the disease and its close relationship with metabolic derangements[16-20]. Given the overall emphasis of this latter association in MAFLD definition (from normal weight to obesity), the new term includes a wide phenotypical range from metabolically unhealthy normal weight to metabolically unhealthy. Nevertheless, an accurate definition of metabolic health is still lacking, especially in patients with obesity[21].

Table 1 Comparison between metabolic associated fatty liver disease and non-alcoholic fatty liver disease diagnostic criteria.
MAFLD criteria[1]
NAFLD criteria[62]
Histological (biopsy), imaging or blood biomarker evidence of hepatic steatosis and the presence of one of these criteria:Presence of steatosis in > 5% of hepatocytes detected by biopsy
(1) Overweight/obesity-The proton density fat fraction (providing a rough estimation of the volume fraction of fatty material in the liver) > 5.6% assessed by proton magnetic resonance spectroscopy
(2) Diabetes mellitus
(3) Evidence of metabolic dysregulation defined as the presence of ≥ 2 of the following conditions:
(a) Waist circumference ≥ 102 cm in Caucasian men and 88 cm in women (or ≥ 90/80 cm in Asian men and women);
(b) Blood pressure ≥ 130/85 mmHg or specific drug treatment; (c) triglyceride ≥ 1.70 mmol/L or specific drug treatment;
(d) High-density lipoprotein cholesterol < 1.0 mmol/L for men and < 1.3 mmol/L for women;
(e) Prediabetes (i.e., fasting glucose levels 5.6–6.9 mmol/L, or 2-h postload glucose levels 7.8–11.0 mmol/L or hemoglobin A1c 5.7%–6.4%;
(f) Homeostasis model assessment-insulin resistance score ≥ 2.5;
and (g) High sensitive C-reactive protein > 2 mg/L
-Quantitative fat/water selective magnetic resonance imaging
Exclusion of both secondary causes and a daily alcohol consumption ≥ 30 g for men and 20 g for women
Table 2 Main findings of the studies on MAFLD genetics.
Gene
Study design
Population
Gene pathophysiology
Main findings                     
b-Klotho (KLB) genePanera et al[57], Hospital-based retrospective cohort study1111 adult Italian MAFLD patients from the Metabolic Liver Diseases outpatient service at Fondazione IRCCS Ca’Granda of Milan between January 1999 and December 2019. Patients were stratified according to obesity status:The rs17618244 G>A variant in the b-Klotho (KLB) gene encodes for a transmembrane protein which complexes with Fibroblast Growth Factor Receptors to bind the hormones FGF21 and FGF19. Both genes play an important role in lipid and glucose metabolism and in obesityKLB rs17618244 variant was linked to hepatic fibrosis
-BMI > 35: 708 subjectsKLB A allele was associated with lobular inflammation and cirrhosis in patients stratified for obesity status; Hepatic KLB mut expression seemed to be linked to proliferative rate improvement and pro-fibrogenic genes induction
-BMI ≤ 35: 403 subjects
Inclusion criteria were liver biopsy or severe obesity and availability of DNA samples
Hydroxysteroid 17-beta dehydrogenase 13 (HSD17B13) geneLiu et al[59], Cross-sectional analysis427 Han Chinese from the PERSONS cohort with biopsy confirmed MAFLD; Hydroxysteroid 17-beta dehydrogenase 13 (HSD17B13) gene encodes a hepatic lipid droplet proteinData confirmed that the HSD17B13 region is a susceptibility locus for MAFLD-related fibrosis
Aged ≥ 18 yr
An effect of modulated PNPLA3 rs738409 on hepatic steatosis was reported
Significant differences in levels of fasting glucose, triglycerides, and high-density lipoprotein cholesterol among subject with HSD17B13- rs72613567 (TA allele) genotypes were observed, but no differences in biochemical parameters among the rs6531975 (A allele) genotypes were found; The minor TA allele was linked to an increased risk of fibrosis, while the minor A allele had a protective effect against liver damage
Membrane-bound O-acyltransferase domain-containing protein 7 (MBOAT7) (1) Meroni et al[52], Review: 21 studies: -6 case control studies; -10 case only; -2 metanalysis; -2 GWAS; -1 cohort studies (1) Age: -4 pediatric studies; -17 adult studies; Ethnicity: -14 Caucasian; -5 multiethnic; -2 AsianThe MBOAT7 codifies for an enzyme highly expressed in hepatocytes, hepatic stellate cells and hepatic sinusoidal cells; It has been involved in fatty acid metabolism and in hepatic both inflammation and fibrosis (1) In patients with MAFLD, MBOAT7 might affect liver damage
Downregulation of liver expression of MBOAT7 induces changes in phosphoinositide composition pattern with subsequent modified membrane lipid composition and lipid mediator profiles
Hyperinsulinemia, is a cofactor for MBOAT impairment; MBOAT7 dysfunction may influence liver disease progression to steatohepatitis and fibrosis and chronic hyperinsulinemia to steatosis development
(2) Ismaiel et al[53], Review: 22 studies: -7 case control studies; -3 case only; -5 metanalysis; -7 cohort studies (2) A total of 22 studies: -4 pediatric studies with ultrasound (US) diagnosis of fatty liver; -18 adult studies: 17 with fatty liver diagnosis with liver biopsy/ imaging and 1 with US (2) Except for Asian population, studies on European, Hispanic, and African American adults with MAFLD evaluating the rs641738 variant reported a downregulation of the MBOAT7 expression, which increased MAFLD severity, liver fat, NASH progression, advanced fibrosis, and HCC
No association with coronary artery disease was found. In children with obesity this variant was associated with increased plasma ALT levels
Table 3 Metabolic syndrome criteria in adults and children.

Abdominal obesity
Hypertension
Dyslipidemia
Fasting glucose
IDF central obesity + 2 of 4 criteria in adult patients and children aged >10 yr[87-89]10–15 yr old waist circumference (WC) ≥ 90th percentile for age and sex Systolic BP ≥ 130 mmHg or diastolic BP ≥ 85 mmHgTG ≥ 150 mg/dL or specific treatment HDL < 40 mg/dL (male), HDL < 50 mg/dL (female)≥ 100 mg/dL or diagnosis of type 2 diabetes mellitus
>15 yr old WC ≥ 94 cm (male) b WC ≥ 80 cm (female)
Panel: IDEFICS definition of metabolic syndrome in children aged 2–11 yr[90]110–15 yr old WC ≥ 90th percentile for age and sex Blood pressure: systolic ≥ 90th percentile or diastolic ≥ 90th percentile TG: ≥ 90th percentile or HDL cholesterol: ≤ 10th percentileInsulin ≥ 90th percentile or fasting glucose ≥ 90th percentile
> 15 yr old adults criteria

An increasing number of studies have explored metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO) in adult and pediatric cohorts[22-24]. MUO individuals have a higher cardiovascular risk than their metabolically healthy counterparts. However, MHO also might predispose over time to an increased risk of cardiometabolic derangements[25-27]. In light of this, a detailed clinical assessment of the cardiometabolic risk in children (including evaluation of anthropometric measures such as weight, height, waist, and hip circumferences according to age- and gender-specific percentiles and Acanthosis nigricans detection as a clinical marker of IR) represents a crucial first step for the evaluation of these patients.

Adipose distribution pattern is considered to have a critical influence on MAFLD development, as demonstrated by the positive correlation of amount of visceral adipose tissue with liver inflammation and fibrosis[4].

To date, the clinical feasibility of MAFLD definition has been mostly studied in adults, but a similar growing interest is also emerging in children. Therefore, we aimed to provide a comprehensive overview by summarizing the most recent evidence on the tangled puzzle of MAFLD in adults and children.

PATHOPHYSIOLOGY

Fatty liver pathophysiology includes a well-known spectrum of determinants such as inflammation, IR, genetics and environment[4,28,29]. Genetic determinants commonly implied in NAFLD susceptibility (such as PNPLA3[30-32], TM6SF2[33], MBOAT7[34-36] and HSD17B13[37-42] genes) have been also linked to MAFLD pathogenesis[43-45] (Table 2). In particular, the effect of the PNPLA3 I148M polymorphism as a key genetic factor for NAFLD susceptibility across different ethnicities has been largely recognized both in adults and children[45]. Similarly, robust data have also supported the role of the TM6SF2 gene in hepatic steatosis development both in adults and children[46-48]. Noteworthy, a pleiotropic effect has been described for both genes because of their extrahepatic role in affecting also kidney function in children with obesity[49,50] and adult with T2D and fatty liver[51]. In addition, robust evidence showed that the downregulation of the MBOAT7 gene predisposed to fatty liver development both in children and adults[34,52,53]. In contrast, the HSD17B13 variant has been recognized as a protective factor against liver injury and its progression[38,54,55]. As described for other well-known single nucleotide polymorphisms related to fatty liver, this variant has been found also to influence kidney function[56].

Minor genetic variants affecting IR, oxidative stress and inflammation pathways have been found to be related to fatty liver development[45,57]. In particular, a significant association between the rs17618244 G>A variant in the KLB gene and hepatic fibrosis has been described, and this gene is a central player in obesity and lipid and glucose metabolism, as demonstrated by its association with lobular inflammation and cirrhosis in patients clustered according to obesity degree[57].

MAFLD genetic susceptibility is still poorly explored[58,59]. Liu et al[59] confirmed the role of the HSD17B13 region in a cohort of 427 Han Chinese adults as a genetic factor predisposing to MAFLD-related fibrosis and of modulated PNPLA3 rs738409 polymorphism on fatty liver development[58]. Recent evidence supports an inverse allelic effect of the association of HSD17B13 variants on liver damage: in particular, hepatic fibrosis risk has been found to be increased by the minor allele TA of the rs72613567 variant, while a protective role against liver damage for the minor A allele of the rs6531975 variant has been demonstrated[59].

EVIDENCE ON MAFLD: FROM ADULTHOOD TO CHILDHOOD

As the renaming of the liver condition, the clinical usefulness of MAFLD definition has been tested in several studies[60-64] (Table 2). Lin et al[60] first compared MAFLD and NAFLD criteria in a large cohort of 13 083 subjects grouped as MAFLD (31.24%), NAFLD (33.23%) and non-metabolic-dysfunction-associated NAFLD (non-MD-NAFLD) (4.74%) (e.g., subjects with NAFLD but not covered by MAFLD criteria)[61-62]. Authors found that patients with fatty liver were older, more likely to be male, and have worse cardiometabolic and hepatic profile independently of the used criteria[60].

Compared to NAFLD, MAFLD subjects were older (48.39 ± 15.20 years) and presented with higher body mass index (BMI), liver enzymes, and noninvasive liver fibrosis scores. In addition, an increased percentage of metabolic comorbidities (including diabetes, IR and hypertension) was reported in these patients[60]. Patients in the non-MD-NAFLD group were the youngest and presented with a better metabolic profile than those belonging to the MAFLD and NAFLD groups. In this framework, a more accurate identification of patients at higher risk of negative metabolic consequences seemed to be achieved by MAFLD criteria[60].

Conversely, no significant differences for the main clinical and biochemical variables between NAFLD and MAFLD were found in a large cohort of 780 adult patients with biopsy-proven fatty liver diagnosis [55]. Taking into account the alcohol consumption in MAFLD definition, patients with MAFLD with significant alcohol intake showed a worse hepatic profile (characterized by higher steatosis degree and transaminase levels) compared to those with MAFLD only[55].

The usefulness of MAFLD definition has been also examined by Sun et al[65] in a highly selected population such as patients with chronic kidney disease (CKD). Authors demonstrated a better performance of MAFLD diagnostic criteria than NAFLD in identifying patients with CKD[65], as previously found[64]. Of note, a strong and independent relationship of MAFLD and MAFLD with increased liver fibrosis scores with CKD and abnormal albuminuria was described[65].

Recently, differences between NAFLD and MAFLD criteria were tested in a 2-year follow-up Italian study conducted in 221 patients receiving a new diagnosis of celiac disease (CD) as a high-risk condition for fatty liver[66]. Compared to NAFLD, MAFLD definition allowed a better identification of CD patients at risk of disease progression and the coexistence of fibrosis seemed to enhance the occurrence of adverse outcomes in these patients[66].

Yamamura et al[67] compared the diagnostic accuracy of MAFLD and NAFLD in identifying individuals with significant hepatic fibrosis and clarified the influence of mild alcohol consumption (< 20 g/d) on the degree of the hepatic disease in a large cohort of 765 subjects clustered in two groups as NAFLD and MAFLD. Compared to NAFLD, MAFLD criteria provided careful detection of hepatic fibrosis, as reflected by the strong relationship between certain hepatic fibrosis markers and liver stiffness in patients diagnosed with MAFLD[67]. Given that, dysmetabolic patients at high risk of adverse hepatic outcomes were better identified through MAFLD than NAFLD criteria[12,21].

As the well-known relevance of alcohol intake on hepatic fibrosis risk development was not included in MAFLD definition, the authors also examined its influence on fatty liver severity[67]. Patients with MAFLD and alcohol intake of 1–59 g/d were more likely to be male and to have higher fasting blood glucose, serum liver enzymes, creatinine, and uric acid levels than those with MAFLD and no alcohol consumption[67]. Of note, there is no evidence on the potential negative effect of alcohol intake on renal damage risk in MAFLD individuals[67]. Authors concluded that MAFLD presence was an independent risk factor for significant fibrosis (defined by FIB-4 index ≥ 1.3 and liver stiffness ≥ 6.6 kPa using Shear wave elastography), and both MAFLD and mild alcohol intake were associated with increased prevalence of significant fibrosis (25.0% vs 15.5%)[67].

Further data examining the role of alcohol intake in this context[60] demonstrated a better metabolic profile but increased transaminase levels in subjects with MAFLD having a greater alcohol intake compared to those with no alcohol consumption. However, no consensus has been reached on the effect of alcohol in MAFLD, but some noninvasive fibrosis scores have been positively associated with MAFLD and alcohol intake[60].

Despite accumulating data on the impact of MAFLD on liver disease severity[60,65,67], its influence on the potential malignant transformation into hepatocellular cancer has been not evaluated.

Unlike adults, pediatric MAFLD data are limited. Because of the widespread distribution of this hepatic condition in childhood, recent epidemiological data reported a worrying increase of pediatric MAFLD prevalence[68-70].

MAFLD definition has been tested first in adult subjects; therefore, its clinical utility in a pediatric setting is still under investigation, since the fatty liver etiology at this stage[71-73] and the obesity status[21]. A recent Italian study investigated the usefulness of MAFLD criteria in 954 children with obesity[21]. The authors grouped their cohort as subjects with (1) obesity only; (2) obesity and NAFLD; and (3) obesity, NAFLD and metabolic dysregulation. The latter group was significantly older and showed higher BMI, systolic blood pressure, diastolic blood pressure, waist/hip ratio, HOMA-IR, triglyceride levels, baseline and 2-h oral glucose tolerance test glycemia, and transaminase levels. A higher prevalence of carriers of the PNPLA3 rare allele was reported in this group compared with others. Taken together, these findings suggest a worse cardiometabolic profile in subjects with obesity, fatty liver, and metabolic dysregulation than in those belonging to other groups. As a preliminary study, MAFLD diagnosis based on metabolic dysregulation in children with obesity seemed more accurate for cardiometabolic risk stratification in a high-risk population such as children with obesity[21]. PNPLA3 gene seems to play a role in a wider metabolic milieu beyond NAFLD[21], as previously found in a similar pediatric cohort[50,74].

More recently, an international panel[75] has proposed an age-appropriate MAFLD definition based on sex and age percentiles. Diagnostic criteria for pediatric MAFLD are based on the presence of hepatic steatosis (detected either by liver histology, imaging, blood biomarkers or blood scores) in addition to one of the following conditions: excess adiposity, T2D or prediabetes, or evidence of metabolic dysregulation (defined by the presence of at least two metabolic risk conditions according to sex and age percentiles such as hypertension, increased waist circumference, hypertriglyceridemia, low serum HDL cholesterol levels, triglyceride-to-HDL ratio ≥ 2.25, and impaired fasting glucose)[75].

Contrary to the adult findings, the natural history of fatty liver in children is still not fully understood but its increase has been mainly linked to obesity[75]. Pediatric fatty liver usually does not occur in children < 3 years and is rare in those aged < 10 years. To date, it has been demonstrated that the entire spectrum of liver disease severity (from simple steatosis to steatohepatitis, fibrosis, and end-stage cirrhosis) might occur also in pediatric patients diagnosed with fatty liver, and that the progression is strongly related to IR severity[75]. As a consequence, the occurrence of severe complications (including liver transplantation) at this early age has also been reported. The pivotal role of primary care for early detection of pediatric fatty liver is widely recognized, and lifestyle modifications are the only valid treatment for the disease[75]. Therefore, redefinition of pediatric MAFLD represents a crucial step for global management improvement, including risk stratification and multidisciplinary care.

MAFLD: NEW INSIGHTS AND FUTURE DIRECTIONS

The tangled and multifactorial physiopathology of MAFLD (including inflammation, sex, age, ethnicity, diet and microbiota, hormones, and genetics) is still poorly defined. Despite the centrality of metabolic dysfunction, diagnosing fatty liver is also essential for MAFLD definition. Liver biopsy represents the common diagnostic gold standard for hepatic fat content assessment, but its invasiveness has limited its clinical utility in children[76,77]. A growing number of studies has evaluated different noninvasive biomarkers for MAFLD diagnosis, by identifying novel attractive therapeutic options for the management of the disease[78-81]. In this context, investigation of the gut–liver axis has attracted scientific attention[81-84]. Considering the relevance of the intestinal barrier in multiple biological mechanisms and the crucial influence of the immune system (located in the liver, intestine and adipose tissue)[84], this term strengthens the association of the liver with the gut barrier.

The association of gut–liver axis changes with MAFLD pathophysiology have recently been explored[78], by pointing out the role of inflammation and release of chemokines and cytokines by liver-infiltrating macrophages as key factors for progressive forms of fatty liver[78].

Dysbiosis and gut barrier changes have both been linked to inflammation and metabolic abnormalities in MAFLD. Remarkably, a peculiar association of microbiome alterations with carbohydrates, lipids and amino acids metabolism in MAFLD has also been described[81], but no consensus has been reached in this field. Nevertheless, promising preclinical studies[81] have enriched the spectrum of potential MAFLD therapeutic tools such as fecal microbiota transplantation[82-84]. A similar study on MAFLD adults[84] investigated microbiota-derived metabolites as potential noninvasive biomarkers for MAFLD, by identifying certain metabolites [e.g., phosphatidylcholine (PC), lysoPC, plasma eicosanoic acid or fatty acid 20:1 (FA20:1), PCaaC24:0, xanthine, and triglycerides] as early microbiota-related products involved in liver disease progression[84]. In addition, a significant association of the PNPLA3 gene with plasma monounsaturated fatty acid FA(20:1) or eicosanoic acid was also demonstrated.

Notably, serum mi-RNA-122 (as the major hepatic mi-RNA involved in metabolic diseases) is significantly related to MAFLD progression in subjects with obesity and MAFLD[80]; therefore suggesting their potential prognostic utility for liver disease progression[80].

Although preliminary, some promising evidence supports the identification of novel potential therapeutic targets for MAFLD[85-88]. In particular, a significant decrease in MAFLD prevalence has been reported in normal-weight adolescents treated with a low-dose combination of spironolactone, pioglitazone and metformin (SPIOMET)[86-90] than those with classical hormone therapy, by underlining the role of SPIOMET treatment as a promising new pathophysiological approach in MAFLD patients[88]. Due to the relevant cardiometabolic burden of MAFLD and the absence of effective pharmacological agents both in children and adults, further studies are needed to identify specific noninvasive markers able to improve the management of MAFLD patients[75]. Several novel therapeutic targets based on molecular pathways are under investigation[78,84], but there are no current licensed MAFLD treatments[75].

CONCLUSIONS

The natural history of pediatric MAFLD remains to be defined, but mounting evidence from adults supports a significant increased cardiovascular risk in view of the concomitant occurrence of metabolic impairments with liver disease. Therefore, better knowledge of the intricate MAFLD pathophysiology might pave the way for new therapeutic approaches to improve the management of these patients at greater cardiometabolic risk.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Pediatrics

Country/Territory of origin: Italy

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C, C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Chen GX, China; Chen F, China; Gao Y, China; Ulasoglu C, Turkey; Zhang LL, China S-Editor: Ma YJ L-Editor: Kerr C P-Editor: Ma YJ

References
1.  Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, Zelber-Sagi S, Wai-Sun Wong V, Dufour JF, Schattenberg JM, Kawaguchi T, Arrese M, Valenti L, Shiha G, Tiribelli C, Yki-Järvinen H, Fan JG, Grønbæk H, Yilmaz Y, Cortez-Pinto H, Oliveira CP, Bedossa P, Adams LA, Zheng MH, Fouad Y, Chan WK, Mendez-Sanchez N, Ahn SH, Castera L, Bugianesi E, Ratziu V, George J. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J Hepatol. 2020;73:202-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1239]  [Cited by in F6Publishing: 1917]  [Article Influence: 479.3]  [Reference Citation Analysis (0)]
2.  Morandi A, Di Sessa A, Zusi C, Umano GR, El Mazloum D, Fornari E, Miraglia Del Giudice E, Targher G, Maffeis C. Nonalcoholic Fatty Liver Disease and Estimated Insulin Resistance in Obese Youth: A Mendelian Randomization Analysis. J Clin Endocrinol Metab. 2020;105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
3.  Manco M. Insulin Resistance and NAFLD: A Dangerous Liaison beyond the Genetics. Children (Basel). 2017;4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
4.  Eslam M, Sanyal AJ, George J; International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology. 2020;158:1999-2014.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1188]  [Cited by in F6Publishing: 1586]  [Article Influence: 396.5]  [Reference Citation Analysis (1)]
5.  Kuchay MS, Misra A. From non-alcoholic fatty liver disease (NAFLD) to metabolic-associated fatty liver disease (MAFLD): A journey over 40 years. Diabetes Metab Syndr. 2020;14:695-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
6.  Tilg H, Effenberger M. From NAFLD to MAFLD: when pathophysiology succeeds. Nat Rev Gastroenterol Hepatol. 2020;17:387-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 138]  [Article Influence: 34.5]  [Reference Citation Analysis (0)]
7.  Utzschneider KM, Kahn SE. Review: The role of insulin resistance in nonalcoholic fatty liver disease. J Clin Endocrinol Metab. 2006;91:4753-4761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 555]  [Cited by in F6Publishing: 587]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
8.  Di Bonito P, Valerio G, Licenziati MR, Miraglia Del Giudice E, Baroni MG, Morandi A, Maffeis C, Campana G, Spreghini MR, Di Sessa A, Morino G, Crinò A, Chiesa C, Pacifico L, Manco M. High uric acid, reduced glomerular filtration rate and non-alcoholic fatty liver in young people with obesity. J Endocrinol Invest. 2020;43:461-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
9.  Targher G, Day CP, Bonora E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N Engl J Med. 2010;363:1341-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1326]  [Cited by in F6Publishing: 1361]  [Article Influence: 97.2]  [Reference Citation Analysis (0)]
10.  Targher G. What's Past Is Prologue: History of Nonalcoholic Fatty Liver Disease. Metabolites. 2020;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
11.  Mantovani A, Petracca G, Beatrice G, Tilg H, Byrne CD, Targher G. Non-alcoholic fatty liver disease and risk of incident diabetes mellitus: an updated meta-analysis of 501 022 adult individuals. Gut. 2021;70:962-969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 205]  [Article Influence: 68.3]  [Reference Citation Analysis (0)]
12.  Xian YX, Weng JP, Xu F. MAFLD vs. NAFLD: shared features and potential changes in epidemiology, pathophysiology, diagnosis, and pharmacotherapy. Chin Med J (Engl). 2020;134:8-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 49]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
13.  Cox AJ, West NP, Cripps AW. Obesity, inflammation, and the gut microbiota. Lancet Diabetes Endocrinol. 2015;3:207-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 465]  [Cited by in F6Publishing: 518]  [Article Influence: 57.6]  [Reference Citation Analysis (0)]
14.  Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract. 2014;105:141-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1141]  [Cited by in F6Publishing: 1211]  [Article Influence: 121.1]  [Reference Citation Analysis (0)]
15.  Saltiel AR, Olefsky JM. Inflammatory mechanisms linking obesity and metabolic disease. J Clin Invest. 2017;127:1-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 894]  [Cited by in F6Publishing: 1127]  [Article Influence: 161.0]  [Reference Citation Analysis (0)]
16.  Alberti KG, Zimmet P, Shaw J. Metabolic syndrome--a new world-wide definition. A Consensus Statement from the International Diabetes Federation. Diabet Med. 2006;23:469-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3852]  [Cited by in F6Publishing: 3946]  [Article Influence: 219.2]  [Reference Citation Analysis (0)]
17.  Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA. 2001;285:2486-2497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20476]  [Cited by in F6Publishing: 20023]  [Article Influence: 870.6]  [Reference Citation Analysis (0)]
18.  Zimmet P, Alberti G, Kaufman F, Tajima N, Silink M, Arslanian S, Wong G, Bennett P, Shaw J, Caprio S; International Diabetes Federation Task Force on Epidemiology and Prevention of Diabetes. The metabolic syndrome in children and adolescents. Lancet. 2007;369:2059-2061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 606]  [Cited by in F6Publishing: 596]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
19.  Chiarelli F, Mohn A. Early diagnosis of metabolic syndrome in children. Lancet Child Adolesc Health. 2017;1:86-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
20.  Ahrens W, Moreno LA, Mårild S, Molnár D, Siani A, De Henauw S, Böhmann J, Günther K, Hadjigeorgiou C, Iacoviello L, Lissner L, Veidebaum T, Pohlabeln H, Pigeot I; IDEFICS consortium. Metabolic syndrome in young children: definitions and results of the IDEFICS study. Int J Obes (Lond). 2014;38 Suppl 2:S4-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 193]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
21.  Di Sessa A, Guarino S, Umano GR, Arenella M, Alfiero S, Quaranta G, Miraglia Del Giudice E, Marzuillo P. MAFLD in Obese Children: A Challenging Definition. Children (Basel). 2021;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
22.  Genovesi S, Antolini L, Orlando A, Gilardini L, Bertoli S, Giussani M, Invitti C, Nava E, Battaglino MG, Leone A, Valsecchi MG, Parati G. Cardiovascular Risk Factors Associated With the Metabolically Healthy Obese (MHO) Phenotype Compared to the Metabolically Unhealthy Obese (MUO) Phenotype in Children. Front Endocrinol (Lausanne). 2020;11:27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
23.  Di Bonito P, Miraglia Del Giudice E, Chiesa C, Licenziati MR, Manco M, Franco F, Tornese G, Baroni MG, Morandi A, Maffeis C, Pacifico L, Valerio G; CARITALY Study on the behalf of the Childhood Obesity Study Group of the Italian Society of Pediatric Endocrinology and Diabetology. Preclinical signs of liver and cardiac damage in youth with metabolically healthy obese phenotype. Nutr Metab Cardiovasc Dis. 2018;28:1230-1236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
24.  Chiesa C, Pacifico L, Xi B, Cadenas-Sanchez C. Editorial: Metabolically Healthy and Unhealthy Obese Children and Adolescents. Front Endocrinol (Lausanne). 2020;11:613703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
25.  Eckel N, Li Y, Kuxhaus O, Stefan N, Hu FB, Schulze MB. Transition from metabolic healthy to unhealthy phenotypes and association with cardiovascular disease risk across BMI categories in 90 257 women (the Nurses' Health Study): 30 year follow-up from a prospective cohort study. Lancet Diabetes Endocrinol. 2018;6:714-724.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 229]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
26.  Caleyachetty R, Thomas GN, Toulis KA, Mohammed N, Gokhale KM, Balachandran K, Nirantharakumar K. Metabolically Healthy Obese and Incident Cardiovascular Disease Events Among 3.5 Million Men and Women. J Am Coll Cardiol. 2017;70:1429-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 274]  [Cited by in F6Publishing: 313]  [Article Influence: 44.7]  [Reference Citation Analysis (0)]
27.  Lassale C, Tzoulaki I, Moons KGM, Sweeting M, Boer J, Johnson L, Huerta JM, Agnoli C, Freisling H, Weiderpass E, Wennberg P, van der A DL, Arriola L, Benetou V, Boeing H, Bonnet F, Colorado-Yohar SM, Engström G, Eriksen AK, Ferrari P, Grioni S, Johansson M, Kaaks R, Katsoulis M, Katzke V, Key TJ, Matullo G, Melander O, Molina-Portillo E, Moreno-Iribas C, Norberg M, Overvad K, Panico S, Quirós JR, Saieva C, Skeie G, Steffen A, Stepien M, Tjønneland A, Trichopoulou A, Tumino R, van der Schouw YT, Verschuren WMM, Langenberg C, Di Angelantonio E, Riboli E, Wareham NJ, Danesh J, Butterworth AS. Separate and combined associations of obesity and metabolic health with coronary heart disease: a pan-European case-cohort analysis. Eur Heart J. 2018;39:397-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 173]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
28.  Tarantino G. NAFLD or MAFLD: That is the conundrum. Hepatobiliary Pancreat Dis Int. 2022;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
29.  Brunt EM, Wong VW, Nobili V, Day CP, Sookoian S, Maher JJ, Bugianesi E, Sirlin CB, Neuschwander-Tetri BA, Rinella ME. Nonalcoholic fatty liver disease. Nat Rev Dis Primers. 2015;1:15080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 466]  [Cited by in F6Publishing: 523]  [Article Influence: 58.1]  [Reference Citation Analysis (0)]
30.  Dongiovanni P, Donati B, Fares R, Lombardi R, Mancina RM, Romeo S, Valenti L. PNPLA3 I148M polymorphism and progressive liver disease. World J Gastroenterol. 2013;19:6969-6978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 170]  [Cited by in F6Publishing: 176]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
31.  Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, Boerwinkle E, Cohen JC, Hobbs HH. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2008;40:1461-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2233]  [Cited by in F6Publishing: 2314]  [Article Influence: 144.6]  [Reference Citation Analysis (0)]
32.  Tang S, Zhang J, Mei TT, Guo HQ, Wei XH, Zhang WY, Liu YL, Liang S, Fan ZP, Ma LX, Lin W, Liu YR, Qiu LX, Yu HB. Association of PNPLA3 rs738409 G/C gene polymorphism with nonalcoholic fatty liver disease in children: a meta-analysis. BMC Med Genet. 2020;21:163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
33.  Kozlitina J, Smagris E, Stender S, Nordestgaard BG, Zhou HH, Tybjærg-Hansen A, Vogt TF, Hobbs HH, Cohen JC. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2014;46:352-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 724]  [Cited by in F6Publishing: 821]  [Article Influence: 82.1]  [Reference Citation Analysis (0)]
34.  Mancina RM, Dongiovanni P, Petta S, Pingitore P, Meroni M, Rametta R, Borén J, Montalcini T, Pujia A, Wiklund O, Hindy G, Spagnuolo R, Motta BM, Pipitone RM, Craxì A, Fargion S, Nobili V, Käkelä P, Kärjä V, Männistö V, Pihlajamäki J, Reilly DF, Castro-Perez J, Kozlitina J, Valenti L, Romeo S. The MBOAT7-TMC4 Variant rs641738 Increases Risk of Nonalcoholic Fatty Liver Disease in Individuals of European Descent. Gastroenterology. 2016;150:1219-1230.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 441]  [Article Influence: 55.1]  [Reference Citation Analysis (0)]
35.  Di Sessa A, Umano GR, Cirillo G, Del Prete A, Iacomino R, Marzuillo P, Del Giudice EM. The Membrane-bound O-Acyltransferase7 rs641738 Variant in Pediatric Nonalcoholic Fatty Liver Disease. J Pediatr Gastroenterol Nutr. 2018;67:69-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
36.  Umano GR, Caprio S, Di Sessa A, Chalasani N, Dykas DJ, Pierpont B, Bale AE, Santoro N. The rs626283 Variant in the MBOAT7 Gene is Associated with Insulin Resistance and Fatty Liver in Caucasian Obese Youth. Am J Gastroenterol. 2018;113:376-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
37.  Su W, Mao Z, Liu Y, Zhang X, Zhang W, Gustafsson JA, Guan Y. Role of HSD17B13 in the liver physiology and pathophysiology. Mol Cell Endocrinol. 2019;489:119-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
38.  Abul-Husn NS, Cheng X, Li AH, Xin Y, Schurmann C, Stevis P, Liu Y, Kozlitina J, Stender S, Wood GC, Stepanchick AN, Still MD, McCarthy S, O'Dushlaine C, Packer JS, Balasubramanian S, Gosalia N, Esopi D, Kim SY, Mukherjee S, Lopez AE, Fuller ED, Penn J, Chu X, Luo JZ, Mirshahi UL, Carey DJ, Still CD, Feldman MD, Small A, Damrauer SM, Rader DJ, Zambrowicz B, Olson W, Murphy AJ, Borecki IB, Shuldiner AR, Reid JG, Overton JD, Yancopoulos GD, Hobbs HH, Cohen JC, Gottesman O, Teslovich TM, Baras A, Mirshahi T, Gromada J, Dewey FE. A Protein-Truncating HSD17B13 Variant and Protection from Chronic Liver Disease. N Engl J Med. 2018;378:1096-1106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 484]  [Article Influence: 80.7]  [Reference Citation Analysis (0)]
39.  Anstee QM, Darlay R, Cockell S, Meroni M, Govaere O, Tiniakos D, Burt AD, Bedossa P, Palmer J, Liu YL, Aithal GP, Allison M, Yki-Järvinen H, Vacca M, Dufour JF, Invernizzi P, Prati D, Ekstedt M, Kechagias S, Francque S, Petta S, Bugianesi E, Clement K, Ratziu V, Schattenberg JM, Valenti L, Day CP, Cordell HJ, Daly AK; EPoS Consortium Investigators. Genome-wide association study of non-alcoholic fatty liver and steatohepatitis in a histologically characterised cohort. J Hepatol. 2020;73:505-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 242]  [Article Influence: 60.5]  [Reference Citation Analysis (0)]
40.  Ma Y, Karki S, Brown PM, Lin DD, Podszun MC, Zhou W, Belyaeva OV, Kedishvili NY, Rotman Y. Characterization of essential domains in HSD17B13 for cellular localization and enzymatic activity. J Lipid Res. 2020;61:1400-1409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
41.  Dong XC. A closer look at the mysterious HSD17B13. J Lipid Res. 2020;61:1361-1362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
42.  Tang S, Zhang J, Mei TT, Zhang WY, Zheng SJ, Yu HB. Association of HSD17B13 rs72613567: TA allelic variant with liver disease: review and meta-analysis. BMC Gastroenterol. 2021;21:490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
43.  Krawczyk M, Rau M, Schattenberg JM, Bantel H, Pathil A, Demir M, Kluwe J, Boettler T, Lammert F, Geier A; NAFLD Clinical Study Group. Combined effects of the PNPLA3 rs738409, TM6SF2 rs58542926, and MBOAT7 rs641738 variants on NAFLD severity: a multicenter biopsy-based study. J Lipid Res. 2017;58:247-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 128]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
44.  Cobbina E, Akhlaghi F. Non-alcoholic fatty liver disease (NAFLD) - pathogenesis, classification, and effect on drug metabolizing enzymes and transporters. Drug Metab Rev. 2017;49:197-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 350]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
45.  Lin YC, Wu CC, Ni YH. New Perspectives on Genetic Prediction for Pediatric Metabolic Associated Fatty Liver Disease. Front Pediatr. 2020;8:603654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
46.  Xue WY, Zhang L, Liu CM, Gao Y, Li SJ, Huai ZY, Dai J, Wang YY. Research progress on the relationship between TM6SF2 rs58542926 polymorphism and non-alcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol. 2022;16:97-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
47.  Dongiovanni P, Petta S, Maglio C, Fracanzani AL, Pipitone R, Mozzi E, Motta BM, Kaminska D, Rametta R, Grimaudo S, Pelusi S, Montalcini T, Alisi A, Maggioni M, Kärjä V, Borén J, Käkelä P, Di Marco V, Xing C, Nobili V, Dallapiccola B, Craxi A, Pihlajamäki J, Fargion S, Sjöström L, Carlsson LM, Romeo S, Valenti L. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology. 2015;61:506-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 369]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
48.  Liu YL, Reeves HL, Burt AD, Tiniakos D, McPherson S, Leathart JB, Allison ME, Alexander GJ, Piguet AC, Anty R, Donaldson P, Aithal GP, Francque S, Van Gaal L, Clement K, Ratziu V, Dufour JF, Day CP, Daly AK, Anstee QM. TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Nat Commun. 2014;5:4309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 399]  [Article Influence: 39.9]  [Reference Citation Analysis (0)]
49.  Marzuillo P, Di Sessa A, Cirillo G, Umano GR, Pedullà M, La Manna A, Guarino S, Miraglia Del Giudice E. Transmembrane 6 superfamily member 2 167K allele improves renal function in children with obesity. Pediatr Res. 2020;88:300-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
50.  Targher G, Mantovani A, Alisi A, Mosca A, Panera N, Byrne CD, Nobili V. Relationship Between PNPLA3 rs738409 Polymorphism and Decreased Kidney Function in Children With NAFLD. Hepatology. 2019;70:142-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
51.  Mantovani A, Taliento A, Zusi C, Baselli G, Prati D, Granata S, Zaza G, Colecchia A, Maffeis C, Byrne CD, Valenti L, Targher G. PNPLA3 I148M gene variant and chronic kidney disease in type 2 diabetic patients with NAFLD: Clinical and experimental findings. Liver Int. 2020;40:1130-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
52.  Meroni M, Longo M, Fracanzani AL, Dongiovanni P. MBOAT7 down-regulation by genetic and environmental factors predisposes to MAFLD. EBioMedicine. 2020;57:102866.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
53.  Ismaiel A, Dumitrascu DL. Genetic predisposition in metabolic-dysfunction-associated fatty liver disease and cardiovascular outcomes-Systematic review. Eur J Clin Invest. 2020;50:e13331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
54.  Vilar-Gomez E, Pirola CJ, Sookoian S, Wilson LA, Liang T, Chalasani N. The Protection Conferred by HSD17B13 rs72613567 Polymorphism on Risk of Steatohepatitis and Fibrosis May Be Limited to Selected Subgroups of Patients With NAFLD. Clin Transl Gastroenterol. 2021;12:e00400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
55.  Zhang HB, Su W, Xu H, Zhang XY, Guan YF. HSD17B13: A Potential Therapeutic Target for NAFLD. Front Mol Biosci. 2021;8:824776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
56.  Di Sessa A, Umano GR, Cirillo G, Passaro AP, Verde V, Cozzolino D, Guarino S, Marzuillo P, Miraglia Del Giudice E. Pediatric non-alcoholic fatty liver disease and kidney function: Effect of HSD17B13 variant. World J Gastroenterol. 2020;26:5474-5483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
57.  Panera N, Meroni M, Longo M, Crudele A, Valenti L, Bellacchio E, Miele L, D'Oria V, Paolini E, Maggioni M, Fracanzani AL, Alisi A, Dongiovanni P. The KLB rs17618244 gene variant is associated with fibrosing MAFLD by promoting hepatic stellate cell activation. EBioMedicine. 2021;65:103249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
58.  Wang P, Wu CX, Li Y, Shen N. HSD17B13 rs72613567 protects against liver diseases and histological progression of nonalcoholic fatty liver disease: a systematic review and meta-analysis. Eur Rev Med Pharmacol Sci. 2020;24:8997-9007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
59.  Liu WY, Eslam M, Zheng KI, Ma HL, Rios RS, Lv MZ, Li G, Tang LJ, Zhu PW, Wang XD, Byrne CD, Targher G, George J, Zheng MH. Associations of Hydroxysteroid 17-beta Dehydrogenase 13 Variants with Liver Histology in Chinese Patients with Metabolic-associated Fatty Liver Disease. J Clin Transl Hepatol. 2021;9:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
60.  Lin S, Huang J, Wang M, Kumar R, Liu Y, Liu S, Wu Y, Wang X, Zhu Y. Comparison of MAFLD and NAFLD diagnostic criteria in real world. Liver Int. 2020;40:2082-2089.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 320]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
61.  Alharthi J, Gastaldelli A, Cua IH, Ghazinian H, Eslam M. Metabolic dysfunction-associated fatty liver disease: a year in review. Curr Opin Gastroenterol. 2022;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
62.  Devi J, Raees A, Butt AS. Redefining non-alcoholic fatty liver disease to metabolic associated fatty liver disease: Is this plausible? World J Hepatol. 2022;14:158-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
63.  Kang SH, Cho Y, Jeong SW, Kim SU, Lee JW; Korean NAFLD Study Group. From nonalcoholic fatty liver disease to metabolic-associated fatty liver disease: Big wave or ripple? Clin Mol Hepatol. 2021;27:257-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 58]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
64.  Kawaguchi T, Tsutsumi T, Nakano D, Torimura T. MAFLD: Renovation of clinical practice and disease awareness of fatty liver. Hepatol Res. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 45]  [Article Influence: 22.5]  [Reference Citation Analysis (1)]
65.  Sun DQ, Jin Y, Wang TY, Zheng KI, Rios RS, Zhang HY, Targher G, Byrne CD, Yuan WJ, Zheng MH. MAFLD and risk of CKD. Metabolism. 2021;115:154433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 152]  [Article Influence: 50.7]  [Reference Citation Analysis (0)]
66.  Rispo A, Imperatore N, Guarino M, Tortora R, Alisi A, Cossiga V, Testa A, Ricciolino S, Fiorentino A, Morisco F. Metabolic-associated fatty liver disease (MAFLD) in coeliac disease. Liver Int. 2021;41:788-798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
67.  Qu W, Ma T, Cai J, Zhang X, Zhang P, She Z, Wan F, Li H. Liver Fibrosis and MAFLD: From Molecular Aspects to Novel Pharmacological Strategies. Front Med (Lausanne). 2021;8:761538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
68.  Lee H, Lee YH, Kim SU, Kim HC. Metabolic Dysfunction-Associated Fatty Liver Disease and Incident Cardiovascular Disease Risk: A Nationwide Cohort Study. Clin Gastroenterol Hepatol. 2021;19:2138-2147.e10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 217]  [Article Influence: 72.3]  [Reference Citation Analysis (0)]
69.  Ramírez-Mejía MM, Díaz-Orozco LE, Barranco-Fragoso B, Méndez-Sánchez N. A Review of the Increasing Prevalence of Metabolic-Associated Fatty Liver Disease (MAFLD) in Children and Adolescents Worldwide and in Mexico and the Implications for Public Health. Med Sci Monit. 2021;27:e934134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
70.  Flisiak-Jackiewicz M, Bobrus-Chociej A, Wasilewska N, Lebensztejn DM. From Nonalcoholic Fatty Liver Disease (NAFLD) to Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD)-New Terminology in Pediatric Patients as a Step in Good Scientific Direction? J Clin Med. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
71.  Zimmet P, Alberti KG, Kaufman F, Tajima N, Silink M, Arslanian S, Wong G, Bennett P, Shaw J, Caprio S; IDF Consensus Group. The metabolic syndrome in children and adolescents - an IDF consensus report. Pediatr Diabetes. 2007;8:299-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1147]  [Cited by in F6Publishing: 1179]  [Article Influence: 69.4]  [Reference Citation Analysis (0)]
72.  Yıldız Y, Sivri HS. Inborn errors of metabolism in the differential diagnosis of fatty liver disease. Turk J Gastroenterol. 2020;31:3-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
73.  Yodoshi T, Orkin S, Arce-Clachar AC, Bramlage K, Xanthakos SA, Valentino PL, Mouzaki M. Alternative Etiologies of Liver Disease in Children With Suspected NAFLD. Pediatrics. 2021;147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
74.  Marzuillo P, Di Sessa A, Guarino S, Capalbo D, Umano GR, Pedullà M, La Manna A, Cirillo G, Miraglia Del Giudice E. Nonalcoholic fatty liver disease and eGFR levels could be linked by the PNPLA3 I148M polymorphism in children with obesity. Pediatr Obes. 2019;14:e12539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
75.  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. Defining paediatric metabolic (dysfunction)-associated fatty liver disease: an international expert consensus statement. Lancet Gastroenterol Hepatol. 2021;6:864-873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 93]  [Article Influence: 31.0]  [Reference Citation Analysis (1)]
76.  Hashimoto E, Taniai M, Tokushige K. Characteristics and diagnosis of NAFLD/NASH. J Gastroenterol Hepatol. 2013;28 Suppl 4:64-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 161]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
77.  Sumida Y, Nakajima A, Itoh Y. Limitations of liver biopsy and non-invasive diagnostic tests for the diagnosis of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. World J Gastroenterol. 2014;20:475-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 370]  [Cited by in F6Publishing: 398]  [Article Influence: 39.8]  [Reference Citation Analysis (2)]
78.  Martín-Mateos R, Albillos A. The Role of the Gut-Liver Axis in Metabolic Dysfunction-Associated Fatty Liver Disease. Front Immunol. 2021;12:660179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 43]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
79.  Sharpton SR, Schnabl B, Knight R, Loomba R. Current Concepts, Opportunities, and Challenges of Gut Microbiome-Based Personalized Medicine in Nonalcoholic Fatty Liver Disease. Cell Metab. 2021;33:21-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 95]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]
80.  Hegazy MA, Abd ALgwad I, Abuel Fadl S, Sayed Hassan M, Ahmed Rashed L, Hussein MA. Serum Micro-RNA-122 Level as a Simple Noninvasive Marker of MAFLD Severity. Diabetes Metab Syndr Obes. 2021;14:2247-2254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
81.  Duan Y, Llorente C, Lang S, Brandl K, Chu H, Jiang L, White RC, Clarke TH, Nguyen K, Torralba M, Shao Y, Liu J, Hernandez-Morales A, Lessor L, Rahman IR, Miyamoto Y, Ly M, Gao B, Sun W, Kiesel R, Hutmacher F, Lee S, Ventura-Cots M, Bosques-Padilla F, Verna EC, Abraldes JG, Brown RS Jr, Vargas V, Altamirano J, Caballería J, Shawcross DL, Ho SB, Louvet A, Lucey MR, Mathurin P, Garcia-Tsao G, Bataller R, Tu XM, Eckmann L, van der Donk WA, Young R, Lawley TD, Stärkel P, Pride D, Fouts DE, Schnabl B. Bacteriophage targeting of gut bacterium attenuates alcoholic liver disease. Nature. 2019;575:505-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 429]  [Article Influence: 85.8]  [Reference Citation Analysis (0)]
82.  Sabino J, Hirten RP, Colombel JF. Review article: bacteriophages in gastroenterology-from biology to clinical applications. Aliment Pharmacol Ther. 2020;51:53-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
83.  Hsu BB, Gibson TE, Yeliseyev V, Liu Q, Lyon L, Bry L, Silver PA, Gerber GK. Dynamic Modulation of the Gut Microbiota and Metabolome by Bacteriophages in a Mouse Model. Cell Host Microbe. 2019;25:803-814.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 272]  [Article Influence: 54.4]  [Reference Citation Analysis (0)]
84.  Mazzini FN, Cook F, Gounarides J, Marciano S, Haddad L, Tamaroff AJ, Casciato P, Narvaez A, Mascardi MF, Anders M, Orozco F, Quiróz N, Risk M, Gutt S, Gadano A, Méndez García C, Marro ML, Penas-Steinhardt A, Trinks J. Plasma and stool metabolomics to identify microbiota derived-biomarkers of metabolic dysfunction-associated fatty liver disease: effect of PNPLA3 genotype. Metabolomics. 2021;17:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
85.  DiVall S, Merjaneh L. Adolescent Polycystic Ovary Syndrome: An Update. Pediatr Ann. 2019;48:e304-e310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
86.  Ibáñez L, Oberfield SE, Witchel S, Auchus RJ, Chang RJ, Codner E, Dabadghao P, Darendeliler F, Elbarbary NS, Gambineri A, Garcia Rudaz C, Hoeger KM, López-Bermejo A, Ong K, Peña AS, Reinehr T, Santoro N, Tena-Sempere M, Tao R, Yildiz BO, Alkhayyat H, Deeb A, Joel D, Horikawa R, de Zegher F, Lee PA. An International Consortium Update: Pathophysiology, Diagnosis, and Treatment of Polycystic Ovarian Syndrome in Adolescence. Horm Res Paediatr. 2017;88:371-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 191]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
87.  Legro RS, Arslanian SA, Ehrmann DA, Hoeger KM, Murad MH, Pasquali R, Welt CK; Endocrine Society. Diagnosis and treatment of polycystic ovary syndrome: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2013;98:4565-4592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1035]  [Cited by in F6Publishing: 984]  [Article Influence: 89.5]  [Reference Citation Analysis (0)]
88.  de Zegher F, López-Bermejo A, Ibáñez L. Central Obesity, Faster Maturation, and 'PCOS' in Girls. Trends Endocrinol Metab. 2018;29:815-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
89.  Ibáñez L, Díaz M, García-Beltrán C, Malpique R, Garde E, López-Bermejo A, de Zegher F. Toward a Treatment Normalizing Ovulation Rate in Adolescent Girls With Polycystic Ovary Syndrome. J Endocr Soc. 2020;4:bvaa032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
90.  de Zegher F, Diaz M, Ibañez L. From adolescent PCOS to adult MAFLD: opposing effects of randomised interventions. BMJ Open Gastroenterol. 2021;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]