Next Article in Journal
Retrospective Analysis of Vitamin D Deficiency in an Adult Population of Arad County, Western Romania (2019–2022)
Previous Article in Journal
Improvement in the Stability and Enzymatic Activity of Pleurotus sapidus Lipoxygenase Dissolved in Natural Deep Eutectic Solvents (NADESs)
Previous Article in Special Issue
Non-Invasive Diagnostic of NAFLD in Type 2 Diabetes Mellitus and Risk Stratification: Strengths and Limitations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NAFLD Fibrosis Progression and Type 2 Diabetes: The Hepatic–Metabolic Interplay

by
Simona Cernea
1,2
1
Department M3, Internal Medicine I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, 540142 Târgu Mureş, Romania
2
Diabetes, Nutrition and Metabolic Diseases Outpatient Unit, Emergency County Clinical Hospital, 540136 Târgu Mureş, Romania
Life 2024, 14(2), 272; https://doi.org/10.3390/life14020272
Submission received: 15 January 2024 / Revised: 13 February 2024 / Accepted: 14 February 2024 / Published: 18 February 2024

Abstract

:
The bidirectional relationship between type 2 diabetes and (non-alcoholic fatty liver disease) NAFLD is indicated by the higher prevalence and worse disease course of one condition in the presence of the other, but also by apparent beneficial effects observed in one, when the other is improved. This is partly explained by their belonging to a multisystemic disease that includes components of the metabolic syndrome and shared pathogenetic mechanisms. Throughout the progression of NAFLD to more advanced stages, complex systemic and local metabolic derangements are involved. During fibrogenesis, a significant metabolic reprogramming occurs in the hepatic stellate cells, hepatocytes, and immune cells, engaging carbohydrate and lipid pathways to support the high-energy-requiring processes. The natural history of NAFLD evolves in a variable and dynamic manner, probably due to the interaction of a variable number of modifiable (diet, physical exercise, microbiota composition, etc.) and non-modifiable (genetics, age, ethnicity, etc.) risk factors that may intervene concomitantly, or subsequently/intermittently in time. This may influence the risk (and rate) of fibrosis progression/regression. The recognition and control of the factors that determine a rapid progression of fibrosis (or its regression) are critical, as the fibrosis stages are associated with the risk of liver-related and all-cause mortality.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide. It has been defined by the presence of steatosis in >5% of hepatocytes, as evidenced by histologic or specific imagistic methods (i.e., magnetic resonance imaging proton density fat fraction (MRI-PDFF) or magnetic resonance spectroscopy), in the absence of secondary causes of liver steatosis (such as excessive alcohol use, viral or autoimmune hepatitis, drugs, etc.) [1,2]. It comprises a spectrum of histopathological and clinical conditions, ranging from simple steatosis (±mild inflammation or ballooning, without evidence of hepatocyte injury) (non-alcoholic fatty liver, NAFL) to non-alcoholic steatohepatitis (NASH) (characterized by steatosis with inflammation and ballooning ± various degrees of fibrosis that can progress to liver cirrhosis) and hepatocellular carcinoma (HCC) [1,3].
The condition is viewed as part of a multisystemic disease, as it is frequently associated with components of the metabolic syndrome (type 2 diabetes mellitus (T2DM), obesity, and dyslipidemia) and cardio-renal comorbidities (cardiovascular disease, atrial fibrillation, chronic kidney disease, etc.) [4]. In fact, it has been considered the hepatic manifestation of the metabolic syndrome, with insulin resistance being a core pathogenetic mechanism [5,6]. In line with this, a panel of international experts proposed in 2020 a new terminology and definition criteria, which are more inclusive and homogenous than NAFLD [7]. The metabolic dysfunction-associated fatty liver disease (MAFLD) was defined as evidence of hepatic steatosis, associated with at least one of the following: overweight/obesity, T2DM, or metabolic dysregulation [7]. More recently, in June 2023, three scientific societies (the European Association for the Study of the Liver (EASL), the American Association for the Study of Liver Diseases (AASLD), and La Asociación Latinoamericana para el Estudio del Hígado (ALEH)) agreed upon the change in nomenclature to metabolic dysfunction-associated steatotic liver disease (MASLD) to reduce stigma and use affirmative criteria and definition instead, and the new nomenclature was endorsed by other professional organizations and societies [8,9]. MASLD was defined as hepatic steatosis with at least one of five cardiometabolic risk factors (in adults): body mass index (BMI) ≥ 25 kg/m2 or waist circumference ≥ 94/80 cm in Caucasian men/women (or ethnicity adjusted); fasting serum glucose ≥ 100 mg/dL, 2 h post-load glucose ≥ 140 mg/dL, glycated hemoglobin (HbA1c) ≥ 5.7%, T2DM, or treatment for T2DM; blood pressure ≥ 130/85 mmHg or specific drug treatment; plasma triglycerides ≥ 150 mg/dL or specific drug treatment; plasma high-density lipoprotein (HDL) cholesterol < 40 mg/dL for men and <50 mg/dL for women or specific drug treatment), in the absence of other causes [8]. Insulin resistance remains the key metabolic dysfunction underlying MASLD [8]. The new nomenclature is thought to bring a better understanding of the disease and increased awareness, as well as support for further biomarkers and drug development [8]. Thus, the changes refer not only to the terms, but also to the definition, since for MASLD the presence of a cardio-metabolic risk factor are required.
In this narrative review, we will discuss the factors associated with the presence and progression of NAFLD-related fibrosis, with special emphasis on the relationship with T2DM, and while transitioning to the new nomenclature, the terms NAFLD/NASH will still be used here.

2. NAFLD and T2DM Bidirectional Relationship

The significant burden of the disease is mirrored by its high global prevalence, as well as its hepatic and extrahepatic consequences. It is estimated that NAFLD affects about a quarter of the adult population worldwide, and slightly more than half of patients with T2DM, with some geographic differences [10,11,12]. More significant is the fact that the advanced stages of the disease are present in a larger proportion of patients with T2DM. Overall, the prevalence of NASH in the general population is about 2–6% worldwide, while in patients with T2DM it is 20–40% [10,11,12,13]. Similarly, the prevalence of advanced fibrosis (F2–4) was reported to be around 5–7% in the general population, and 12–20% in patients with T2DM [10,11,12,13,14,15,16]. Moreover, the risk of HCC is 2.0–2.5 fold higher in subjects with T2DM [17,18,19,20]. A recent individual participant-level data meta-analysis of six retrospective studies (2016 participants with NAFLD) indicated that T2DM was an independent predictor of incident HCC (adjusted hazard ratio (HR): 5.34 [1.67–17.09], p = 0.0048) and of incident hepatic decompensation (adjusted HR: 2.15 [1.39–3.34], p = 0.0006) [21].
Diabetes worsens the course of the disease, and in fact, histopathologic analyses of biopsy-proven NAFLD samples demonstrated that patients with T2DM have more advanced fibrosis [22,23,24]. There is a large body of literature that indicates T2DM as an important independent predictor and risk factor for advanced liver disease, including advanced fibrosis, cirrhosis, HCC, and liver-related hospital admissions and deaths [14,25,26,27,28,29,30]. The meta-analysis by Jarvis H et al. of 12 population-based cohort studies (22.8 million subjects followed-up for 10 years) has shown that T2DM doubles the risk of severe liver disease events (HR: 2.25 [1.83–2.76], p < 0.001) and increases the risk of fatal liver events by 63% [25]. The real-world study by Alexander M et al. of 18 million European patients has also demonstrated that diabetes predicts liver disease progression [31]. In NAFLD/NASH patients, the strongest association with liver outcomes (cirrhosis or HCC) is seen in patients with diabetes at baseline (HR: 2.3 [1.9–2.78]) [31]. Similar results were reported in the study conducted by Kanwal et al. that included 271,906 patients with NAFLD which were followed-up for 9.3 years [26]. Diabetes was the only metabolic risk factor independently associated with the risk of progression to HCC (adjusted HR: 2.77 [2.03–3.77]), and it also increased the risk of cirrhosis by 89% [26].
This bidirectional relationship between NAFLD and T2DM is further substantiated by the results of the retrospective analysis performed by Colosimo and colleagues (637 patients with T2DM) that evaluated the impact of optimized glycemic control on markers of liver steatosis and fibrosis [32]. The change in HbA1c corelated positively with changes in Fibrosis-4 (FIB-4) (after adjustment for confounders) (R = 0.666, overall model test p = 0.001; ANOVA for change in HbA1c, p = 0.037) and with Fatty liver index (FLI) independent of body mass index (BMI) change [32].
It is apparent though that not all persons with diabetes have the same risk of hepatic steatosis and fibrosis, and this is modulated by insulin resistance. Diabetes is a heterogenous disease and in recent years several novel subtypes have been described by cluster analysis [33]. Apparently, these subtypes have different associations with liver outcomes [33,34,35]. Severe insulin-resistant diabetes (SIRD), characterized by insulin resistance (high homoeostasis model assessment 2-insulin resistance (HOMA2-IR) index) and obesity (high BMI), had the highest prevalence of NAFLD (24.1%) in the study by Ahlqvist et al. [33]. The NAFLD-associated rs10401969 variant of the Transmembrane 6 Superfamily Member 2 (TM6SF2) gene was also associated with SIRD [33]. Zaharia et al., who analyzed data from 1105 participants in the German Diabetes Study with newly diagnosed diabetes, also reported that patients assigned to the SIRD cluster had the highest hepatic lipid content and FLI, as well as the highest estimates of liver fibrosis (NAFLD fibrosis score and aspartate aminotransferase (AST)-to-platelet ratio index (APRI)), and after 5 years of follow-up, the prevalence of hepatic fibrosis remained higher in this cluster [34]. Moreover, the same group showed that SIRD had a higher prevalence of the rs738409 (G) polymorphism of the patatin-like phospholipase domain-containing 3 (PNPLA3) gene, which is associated with increased risk and progression of NAFLD [35].
On the other hand, there are convincing data in the literature coming from several meta-analyses which indicate that NAFLD increases the risk of diabetes by about two-fold [36,37,38]. Importantly, the meta-analysis by Mantovani et al. (33 studies, 501,022 individuals, median follow-up of 5 years) has also indicated that the severity of hepatic fibrosis parallels the risk of incident diabetes, independent of other risk factors such as age, sex, adiposity, or metabolic comorbidities [35]. This was confirmed by several longitudinal biopsy studies showing higher risk of incident T2DM in NAFLD patients with significant (≥F2) or progressive fibrosis [39,40,41].
It appears that the inverse relationship is also valid in the sense that improvement or resolution of NAFLD reduces the risk of T2DM and even favors its remission [42,43,44,45]. The retrospective cohort study by Yamazaki et al. (3074 ultrasonography (US)-evaluated participants followed-up for >10 years) reported a reduced incidence of T2DM by about 70% (multivariate odds ratio (OR): 0.27 [0.12–0.61]) with US-defined NAFLD improvement [42]. The same group later reported that improvement in US-detected hepatic steatosis was associated with a higher chance of T2DM remission (adjusted OR: 3.08 [1.94–4.88]) over a follow-up period of about 2 years [44]. A Chinese prospective cohort study evaluated data from 5671 participants and also suggested a favorable effect of US-defined NAFLD improvement on the risk of progression to new onset T2DM (OR: 0.50 [0.32–0.80]), and on diabetes remission (OR: 2.06 [0.96–4.42]) [45].
Thus, T2DM doubles the risk of NAFLD and significantly increases the risk of progression toward the advanced stages of the disease, while NAFLD doubles the risk of diabetes, mainly when advanced fibrosis is present. Improvement in one condition seems to bring favorable effects to the other, but more data are still needed to further validate these findings.

3. Natural History of NAFLD

The classic view on the natural history of NAFLD is that hepatic steatosis is rather a benign condition with a slow progression, while NASH has in fact the potential to advance to fibrosis and cirrhosis [46]. This dichotomous perspective is challenged by newer histological studies, and it is apparent that the natural course of the disease is rather heterogenous, variable, and dynamic, with some NAFL patients progressing toward more advanced stages of fibrosis, some patients being “slow”, while others “fast” progressors, and some patients presenting regression of fibrosis, even from more advanced stages [3,46,47,48].
A meta-analysis of histological studies (11 studies; 411 patients with paired liver biopsies collected at least one year apart) indicated that hepatic fibrosis progressed in 33.6% of patients, remained stable in 43.1%, while 22.3% of cases presented an improvement in fibrosis stage [49]. The annual fibrosis progression rate was estimated to be 0.14 [0.07–0.21] stages in patients with NASH at baseline (corresponding to one stage progression over 7.1 years), and 0.07 stages [0.02–0.11] in those with steatosis alone (corresponding to one stage of progression over 14.3 years) [49]. A more recent meta-analysis by Pe L et al. that included 54 studies (observational and randomized controlled trials (RCTs), with 26,738 patients with NAFLD diagnosed by liver biopsy or imaging) reported that rates of fibrosis progression were similar between baseline fibrosis stages, and the time to progress by one stage varied between 9.9 and 22.2 years from F0 to F3 [50]. In the same study, it was shown that hepatic steatosis resolution occurred in 21% of subjects with NAFL at baseline after about 4.5 years, while 29% of NASH patients presented resolution of steatohepatitis after a median of 3.5 years [50].
Several meta-analyses of histological data from the placebo arms of the RCTs have evaluated the changes in NASH severity over time and thus provided a valuable insight into the natural history of the disease. The study by Ng CH et al. included 43 RCTs (2649 placebo-treated NASH patients) and showed spontaneous improvements in liver histology over time: the pooled estimate of NASH resolution was 11.65% [7.98–16.71], the two-point NAFLD activity score reduction without worsening of fibrosis was 21.11% [17.24–25.57], and the rate of at least one stage reduction in fibrosis was 18.82% [15.65–22.47] [51]. About 23% of patients presented fibrosis progression of ≥ 1 stage [51]. The network meta-analysis conducted by Penissi et al., which evaluated data from 15 phase 2 and 3 RCTs, reported a pooled estimate rate of NASH resolution (without worsening of fibrosis) of 17% ([12–23%], p < 0.01), and of 21% ([13–31%], p < 0.01) for at least one stage of fibrosis improvement without worsening of NASH [52]. Finally, Ampuero and colleagues reported in their meta-analysis of 27 RCTs a pooled efficacy for NASH resolution of 10% [7,8,9,10,11,12], and 18% [15,16,17,18,19,20,21] for improvement in fibrosis in the placebo arms [53].
However, the evolution of NAFLD/NASH in patients with diabetes is insufficiently investigated so far. A nation-wide cohort study from Italy that included 5030 patients with T2DM evaluated hepatic steatosis using the FLI over a period of 3 years and indicated that about 5% of T2DM patients develop and have remission of liver steatosis each year, respectively, suggesting a dynamic evolution in this patient category as well [54]. More recently, a large multicenter study of 3446 paired-liver biopsied patients with NAFLD evaluated the incidence of fibrosis progression in individuals with or without T2DM [55]. More patients with NAFLD and T2DM presented progression of fibrosis (from stages F0–2 to F3–4) compared to NAFLD subjects without T2DM (26.0% vs. 14.1%, p = 0.008), and similar proportions of patients presented fibrosis regression (from F3–4 to F0–2) in the two groups (27% vs. 22%, p = 0.52) [55]. The adjusted rate of fibrosis progression rate was higher in NAFLD patients with T2DM (+0.23 [0.39] versus +0.16 [0.26] stage/year, p = 0.048), who also had a higher adjusted cumulative incidence of fibrosis progression by at least one stage (24% vs. 20% at 4 years, 60% vs. 50% at 8 years, and 93% vs. 76% after 12 years) [55]. The presence of T2DM at baseline, but not HbA1c, was an independent and significant predictor of fibrosis progression (adjusted HR: 1.69 [1.17–2.43], p = 0.005) [55]. However, the incidence of fibrosis regression by ≥1 stage was similar between the two groups, and T2DM did not emerge as a predictor of fibrosis regression [55]. A Japanese cross-sectional multicenter study (1365 biopsy-proven NAFLD) identified diabetes as a significant risk factor for advanced fibrosis (F3–4) (multivariate OR: 2.387 [1.603–3.553], p < 0.0001) [56].
Thus, the natural history of NAFLD in patients with and without T2DM does not evolve in a straightforward and predictable manner, and this is most probably driven by the interaction of a variable number of risk factors and the complex pathophysiological mechanisms involved.

4. Factors Associated with Fibrosis Progression and Regression

It is apparent that the disease trajectory is highly variable due to interference of a number of multilayered risk factors and therapies that may critically influence disease progression and regression. Identifying the factors associated with fibrosis progression, especially rapid progression toward the advanced stages, and also those associated with fibrosis regression is important, as this might enable improved monitoring strategies and support biomarkers and drug development. This is relevant also because fibrosis stage was proven to be associated with the risk of mortality and is the most significant predictor of outcomes [57,58]. A retrospective study with a mean follow-up period of 20 years (646 biopsy-proven NAFLD patients) had previously shown that fibrosis stage and not NASH increased the risk of mortality (HR: 3.04 [1.94–4.78], p < 0.001 for F3 vs. F0, and 6.53 [3.55–12.03], p < 0.001 for F4 vs. F0) [57]. This was also demonstrated in the meta-analysis by Dulai et al. (five cohort studies; 1495 NAFLD patients, with 17,452 patient years of follow-up) [58]. Compared to F0, higher fibrosis stages exponentially increased the risk of liver-related mortality (rate ratios: 1.41 [0.17–11.95], 9.57 [1.67–54.93], 16.69 [2.92–95.36], and 42.30 [3.51–510.34] from F1 to F4), respectively, and also of all-cause mortality (rate ratios: 1.58 [1.19–2.11], 2.52 [1.85–3.42], 3.48 [2.51–4.83], and 6.40 [4.11–9.95] from F1 to F4) [58]. In addition, the presence of advanced liver fibrosis modifies the clinical management, as it requires periodic surveillance for HCC, and in patients with T2DM it may influence the choices of antihyperglycemic therapeutic agents (mainly in advanced cirrhosis, when insulin might become the only option) [17,59].
A number of risk factors for disease progression have been described (Figure 1). Some of them are modifiable (e.g., diet, physical exercise, microbiota composition, etc.) and others are not (e.g., genetics, age, ethnicity, etc.), and these may act concomitantly, in a synergistic manner, or subsequently/intermittently through the lifetime of NAFLD patients, and thus may influence the risk (and rate) of fibrosis progression.
Genetic and epigenetic factors. The genome-wide association studies (GWAS) have identified several gene loci associated with the risk of NAFLD development and progression [60].
The polymorphism of PNPLA3, which codes adiponutrin, a protein involved in lipid remodeling of hepatic triglycerides, mediates NAFLD risk: the single-nucleotide polymorphism (SNP) rs738409 determines a missense variation (I148M) that disrupts the enzymatic activity interfering with lipid catabolism, and has been shown to associate with liver fibrosis and disease progression [61,62,63]. The PNPLA3 I148M also alters retinol release from the hepatic stellate cells (HSCs), determining a subsequent reduction in the secretion of matrix-modulating enzymes and changing the extracellular matrix remodeling, and this might explain the association with hepatic fibrosis development and progression [60,64]. The meta-analysis by Singal et al. (24 studies with 9915 patients) reported that the PNPLA3 rs738409 SNP is associated with fibrosis severity (OR: 1.32 [1.20–1.45]), and the risk was similar in patients with NAFLD and with other liver disease etiologies [63].
However, the gene–diet interaction seems to be an important factor that modulates the effect of the SNP on the risk of fibrosis [63]. In a study that included 452 non-Hispanic white NAFLD subjects, higher carbohydrate intake was positively associated with higher risk of significant fibrosis (≥F2) (adjusted OR: 1.03, p < 0.01), while higher intakes of n-3 polyunsaturated fatty acids (n-3 PUFAs), methionine, choline, and isoflavones presented an inverse association (adjusted OR of 0.17, 0.32, 0.32, with p < 0.01, and 0.74, p = 0.049, respectively) [65]. The PNPLA3 rs738409 G-allele significantly modulated the relationship between the intakes of the above-mentioned dietary components and fibrosis severity in a dose-dependent manner [65]. Moreover, a recent analysis of data from two independent cohorts (7893 and 46,880 participants, respectively) indicated that the PNPLA3-rs738409-GG had additive effects with metabolic risk factors, such as diabetes and obesity: carriers of this genotype with an indeterminate risk of fibrosis (FIB-4 score between 1.3 and 2.67) and diabetes had a similar incidence rate of cirrhosis as patients with a high risk of fibrosis (FIB-4 > 2.67) [66].
The GWAS also identified the TM6SF2 variant rs58542926 as being associated with NAFLD [67]. TM6SF2 is a transmembrane protein involved in the secretion of VLDL cholesterol, and subsequent studies have associated this SNP with a higher risk of advanced fibrosis in patients with NAFLD, independent of other risk factors [60,68,69]. This was not confirmed in the study by Krawczyk et al., which in turn showed that the membrane-bound O-acyltransferase domain containing 7 (MBOAT7) rs641738 variant was associated with the development of hepatic fibrosis (OR: 1.446, p = 0.046) [70]. A meta-analysis of 42 studies which analyzed data from 1,066,175 participants (9688 with liver biopsies) demonstrated that the rs641738 variant was positively associated with advanced fibrosis in Caucasian adults (OR: 1.22 [1.03–1.45], p = 0.021) [71]. Variants of other genes, such as glucokinase regulatory protein (GCKR), which regulates glucose metabolism and de novo lipogenesis, have been correlated with NAFLD-associated fibrosis [72]. The GCKR rs780094 SNP was independently linked to the severity of liver fibrosis (OR: 2.06 [1.43–2.98], p < 0.001) in an Italian histologic study (366 NAFLD patients) [73].
On the other hand, several gene polymorphisms appear to confer protection from hepatic fibrosis (e.g., Krueppel-like factor (KLF) 6 rs3750861, interleukin (IL) 28B rs12979860, Superoxide dismutase (SOD) 2 rs4880, MER protocol-oncogene, tyrosine kinase (MERTK) rs4374383, etc.) [74]. A high-throughput RNA sequencing approach analyzing 206 samples from a histologically defined NAFLD cohort revealed and validated a gene signature for fibrosis progression, with 25 genes differentially expressed through fibrosis stages F2 to F4, and further analysis at the protein level identified aldo-keto reductase family 1 member B10 (AKR1B10) and growth/differentiation factor 15 (GDF15) concentrations as being strongly correlated with fibrosis stage [75].
A retrospective cohort study from southern Italy including 454 participants with NAFLD and with or without T2DM found no significant differences in the distribution of SNPs between the two patient categories [76]. Patients with T2DM who carried the risk alleles had a higher risk of liver fibrosis and significantly higher liver stiffness [76]. Moreover, a recent longitudinal study (407 T2DM-MASLD patients followed-up for 11 years) showed that combined polymorphisms of the PNPLA3 and TM6SF2 (two or more risk alleles) significantly increased the risk of cirrhosis (OR: 18.48 [6.15–55.58]; p < 0.001) and of cirrhosis complications (OR: 27.20 [5.26–140.62]; p < 0.001) [77]. The same impact of the combination of the risk alleles (common genetic variants of PNPLA3, TM6SF2, and HSD17B13) on the severity of NAFLD was also demonstrated by a large cohort study (110,761 individuals from Denmark and 334,691 individuals from the UK Biobank) [78]. A higher number of risk alleles (higher genetic risk score) progressively increased the risk of cirrhosis (up to 12-fold higher) and of HCC (up to 29-fold higher) in individuals from the general population [78].
Epigenetic modifications have also been shown to modulate liver fibrogenesis and they might in fact explain the variability in disease progression and regression in NAFLD patients with similar gene polymorphisms [74,79]. They have been suggested as potential non-invasive markers of disease progression [60]. A differential DNA methylation of pro- or antifibrogenic genes (i.e., transforming growth factor-β (TGFβ)1, platelet-derived growth factor (PDGF)α, or peroxisome proliferator-activated receptor (PPAR)α and PPARδ), genes encoding matrix molecules or remodeling factors, but also chemokines (CCR7 and CCL5) or factors related to the inflammasome (e.g., signal transducer and activator of transcription (STAT)1, and caspase 1 (CASP1)) has been observed in patients with NAFLD, and this was correlated to the fibrosis stage [80,81,82]. In addition, some emerging data suggest that a dysregulated microRNA (miRNA) expression pattern occurs in NAFLD and may influence disease progression/fibrogenesis (e.g., downregulation of miR-122, miR-331-3p, and miR-30c) [74,83,84]. Also, there are sparce data regarding the role of long non-coding RNAs (lncRNAs) in humans with NAFLD fibrosis [85]. Several lncRNAs (such as nuclear enriched abundant transcript 1 (NEAT1), metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), and plasmacytoma variant translocation 1 (PVT1)) proved to be differentially expressed in fibrotic samples from NAFLD patients [85]. Nevertheless, data connecting the ncRNAs and NAFLD-associated fibrosis are not robust enough, and more studies are still needed in this direction [74].
Ethnicity/race. Data in the literature seem to indicate ethnic disparities related to the prevalence of NAFLD/NASH, but this might be influenced also by genetic or socio-economic factors, lifestyle habits, or other factors [86]. The meta-analysis by Rich et al. of 34 studies (368,569 patients) showed that Hispanics had the highest prevalence of NAFLD and NASH, Blacks had the lowest NAFLD/NASH prevalence, while the percentages were intermediate in Whites [87]. Nevertheless, the prevalence of significant fibrosis was similar among the ethnic/racial groups [87]. NAFLD prevalence seems to be similar in Asian Americans as compared to non-Asian Americans, despite the lower prevalence of obesity in this racial group, while the fibrosis stages were similar, as indicated by data from the National Health and Nutrition Examination Surveys 2017–2018 [88].
Age. Some (but not all) studies observed an inverse U-shaped relationship between age and NAFLD prevalence [89]. However, the meta-analysis by Younossi et al. reported a global NAFLD prevalence consistently increasing throughout age categories [10]. Aging also seems to be associated with more severe liver fibrosis [90,91].
Sex, reproductive, and hormonal status. There are sex-based differences in NAFLD/NASH prevalence and severity, partly related to sex hormones [92]. A recent systematic review and meta-analysis of 54 studies (62,239 for NAFLD analysis, and 6444 for the advanced fibrosis analysis) indicated a lower risk of NAFLD in women compared to men, but a higher risk of advanced fibrosis in those with established NAFLD (mainly after 50 years of age) [93]. In premenopausal women, estrogen seems to exert a protective effect against hepatic steatosis and fibrosis, while high testosterone levels double the risk of NASH fibrosis [92,94]. In a cross-sectional study that included 1782 male subjects with T2DM, total testosterone levels were associated with fibrosis progression (adjusted OR: 0.45 [0.29–0.72], for Q1 vs. Q4, ptrend = 0.001) [95]. In addition, a number of dysregulations of endocrine axes (hypothyroidism, growth hormone deficiency, hypercortisolemia, etc.) are associated with NAFLD/fibrosis development and progression [96,97]. There are some suggestions that thyroid hormone levels are associated with the risk of progressive hepatic fibrosis in patients with T2DM, NAFLD, and normal thyroid function [98].
Diet and alcohol intake. There are sparce high-quality data evaluating the impact of diet or dietary components on liver fibrosis progression in subjects with NAFLD/NASH [17]. Higher caloric intake was shown to be positively associated with NAFLD, but there is not enough solid evidence regarding the correlations with the progression of NASH-associated fibrosis [99]. Nevertheless, a hypocaloric diet (with or without physical activity) leading to weight loss (of >10%) has been shown to improve liver fibrosis [100].
Data from several observational studies indicated that adherence to the Mediterranean diet was associated with a lower risk of hepatic fibrosis [101,102,103]. The Med-Diet score appeared to be negatively correlated with markers of liver fibrosis (N-terminal pro-peptide of type III collagen (PRO-C3)) [104]. Moreover, an intervention study in 144 subjects with moderate or severe NAFLD demonstrated that a low-glycemic-index Mediterranean diet alone or combined with a physical activity program (aerobic with/without resistance exercises) was associated with significant reduction in liver fibrosis (estimated by vibration-controlled elastography) after 90 days [105]. Similar results were shown by a previous smaller study [106]. In addition, a study that included 170 subjects with NAFLD reported that adherence to a healthy dietary pattern (characterized by a high intake of nuts, vegetables, fruits, vegetable oils, low-fat dairy, white meat, coffee, and tea) was associated with a lower risk of liver fibrosis (OR: 0.26 [0.10–0.49]), while the Western dietary pattern (defined by a high intake of refined grains, meat, potatoes, eggs, and soft drinks) was associated with higher risk (OR: 4.21 [1.63–8.31]) [107]. A higher intake of hydrogenated fats, read meat, and soft drinks was associated with an increased risk of fibrosis [107]. Similarly, analysis of data from the Framingham Heart Study and the National Health and Nutrition Examination Survey indicated inverse associations between higher diet quality and liver fibrosis [108]. A small RCT (44 subjects with NAFLD) also demonstrated that adherence to a modified alternate-day calorie restriction diet for eight weeks resulted in a significant decrease in liver fibrosis (SWE score: −0.74 [0.19–1.29], p = 0.01) compared to a habitual diet [109].
Several dietary components have been associated with different risks of hepatic fibrosis in NAFLD patients. For example, dietary intake of myristic acid appeared to be higher in patients with NAFLD fibrosis (p = 0.02), while deficient choline intake was associated with higher liver fibrosis in post-menopausal women with NAFLD (p = 0.002) [110,111]. On the other hand, a meta-analysis of eleven studies demonstrated a protective effect of coffee consumption against liver fibrosis (RR: 0.65 [0.54–0.78], p < 0.00001) [112]. A small 12-week intervention study showed that a low free-sugar diet may reduce hepatic fibrosis in NAFLD subjects with overweight/obesity [113].
Although some cross-sectional studies have suggested that moderate alcohol intake is associated with less severe fibrosis in NAFLD patients, others have shown opposite results, while several longitudinal studies have demonstrated that even a modest alcohol consumption (>10 g/day) or heavy binge drinking may worsen fibrosis and increase the risk of liver-related outcomes [114,115,116,117].
Physical activity and sarcopenia. Several studies have indicated an independent association between NAFLD-related fibrosis and sarcopenia [118,119,120,121]. There are some indications that sarcopenia and/or severe myosteatosis are also associated with fibrosis progression, but more data are needed to substantiate these findings [122,123]. Myokines are suggested to be the mediators of the altered liver–muscle crosstalk, but sarcopenia and NAFLD fibrosis also share some other pathophysiological pathways (i.e., insulin resistance, chronic inflammation, alterations in the regulation of hormone signaling, etc.) that might explain the bidirectional link between them [124,125]. In fact, exercise-induced irisin might mediate the effect of physical activity on NAFLD through its anti-inflammatory and antioxidant properties [126,127]. Indeed, some but not all studies indicated a reduction in liver fibrosis with high-intensity interval training or moderate-to-vigorous aerobic exercise [128,129,130]. Additional research is needed, however, in order to decipher the independent effect of exercise on biopsy-proven NAFLD fibrosis.
Gut microbiota. The altered gut–liver axis has been linked to NAFLD pathogenesis through a number of mechanisms that include the dysbiosis of gut microbiota, which favors disease progression by increasing inflammation, hepatocellular toxicity/damage, and fibrogenesis [131,132]. Gut dysbiosis increases intestinal permeability, favoring toxic bacterial product translocation, higher free fatty acid (FFA) absorption, increased pro-inflammatory cytokines, lipopolysaccharide and ammonia, also intestinal inflammation and dysmotility, etc. [131,133]. Patients with NAFLD have an altered composition of gut microbiota and exhibit a distinct microbial signature, although the microbiome profile is not entirely deciphered [134]. It seems though that there is an increase in the Bacteroidetes and a decrease in the Firmicutes phyla associated with significant liver fibrosis, although other changes have been observed (increase in Proteobacteria phylum and several species such as Bacteroides, Escherichia coli, and possibly Ruminococcus; decrease in Prevotella, etc.) [135,136,137]. Modulation of the gut microbiota through lifestyle intervention might influence the disease/fibrosis progression, as suggested by the subanalysis of the PREDIMED-Plus trial data, but more evidence is needed in this respect [138].
Obesity. Excessive (and dysfunctional) body adiposity, mainly abdominal/visceral obesity, has been linked to the development and progression of NAFLD/liver fibrosis through the release of pro-inflammatory cytokines, FFAs, and adipokines, and hepatic insulin resistance [139,140,141]. Two large cohort studies from Korea (59,957 and 40,700 adults with NAFLD, respectively) showed that obesity/weight gain correlated with worsening of hepatic fibrosis (assessed by non-invasive biomarkers) over a median follow-up period of 7.7 and 6.0 years, respectively [142,143]. Another longitudinal cohort study from Spain (1478 adult subjects) showed that abdominal obesity was associated with moderate-to-advanced liver fibrosis development over a median follow-up of 4.2 years (β: 0.27 [0.11–0.43], p = 0.001) [144]. On the other hand, intervention studies have demonstrated that significant weight loss (≥10% body weight) through lifestyle intervention was associated with the regression of fibrosis in a significant proportion (45%) of NAFLD patients [100]. The analysis of data from two RCTs also demonstrated that for each kg of weight loss, there was a 5% ([2–8%], p = 0.001) increase in the odds of fibrosis improvement [145].
Insulin resistance and type 2 diabetes. As mentioned above, the evidence from the literature points to T2DM as an important metabolic condition associated with the progression of NAFLD-related fibrosis, as well as with a more advanced fibrosis. By using a non-invasive marker of fibrosis (FIB-4) in a sample population of 266 subjects with both NAFLD and T2DM, we have found that about half of them had a significant risk of fibrosis (FIB-4 ≥ 1.3), and 9.4% presented a high risk of advanced fibrosis (FIB-4 ≥ 2.67) (Cernea S., data not published). Moreover, longitudinal studies indicate overall that T2DM is associated with a higher risk of progression of fibrosis [146,147,148]. In addition, a recent case–control study (161 T2DM offsprings and 78 controls) has shown that parental history of T2DM increased the risk of NAFLD-associated significant fibrosis evaluated by transient elastography (OR: 8.89 [1.09–72.01], p = 0.041) after adjustment for confounding factors (age, gender, metabolic traits, and PNPLA3 and TM6SF2 polymorphisms) [149]. Previous research has indicated that parental history of T2DM is associated with a higher risk of NAFLD and higher non-fasting liver enzymes, with body adiposity being a substantial contributing factor [150,151].
Apparently, early glucose derangements (prediabetes, higher glucose variability as evidenced by the continuous glucose monitoring system) and higher post-prandial blood glucose are also correlated with the severity of liver fibrosis, suggesting that hyperglycemia per se might be involved in fibrosis progression [144,152,153,154]. In patients with NAFLD and T2DM, the increase in HbA1c was significantly associated with liver fibrosis progression (standardized coefficient: 0.17 [0.009–0.326], p = 0.038) in a liver biopsy study [155]. The mechanisms behind these associations are not entirely clear, but a gene expression analysis suggested that diabetes may induce hypoxia and oxidative stress in hepatocytes, which may mediate inflammation and fibrosis [155]. In fact, hyperglycemia/glucotoxicity and lipotoxicity induce oxidative stress, mitochondrial dysfunction, and endoplasmic reticulum stress that are involved in the occurrence and progression of NAFLD [156]. Gluco- and lipotoxicity are interrelated and further aggravate insulin resistance [157]. Insulin resistance is, at least in part, the link between T2DM and NAFLD, is involved in the pathogenesis of both conditions, and is a key contributor to NAFLD progression [48]. Moreover, other research indicated that high fasting insulin concentrations or treatment with insulin increased the risk of progression to advanced fibrosis (OR: 1.36, p < 0.001) [158,159]. It has been suggested that insulin is pro-fibrinogenic, as it induces the proliferation of HSCs, the upregulation of connective tissue growth factor, and collagen synthesis [160,161].
On the other hand, antihyperglycemic agents seem to bring benefit in NAFLD, including fibrosis, although more RCTs in biopsy-proven NAFLD patients are still needed for some of the agents. Incretin-based therapies were associated with a decrease in serum alanine aminotransferase (ALT) (−14.1 IU/L [8.3–19.8], p < 0.0001), as indicated by a meta-analysis of four studies (136 participants with NAFLD), with two of them indicating a reduction in liver fibrosis [162]. Two RCTs have shown that treatment with glucagon-like peptide-1 (GLP-1) receptor agonists reduced the progression of hepatic fibrosis [163,164]. A meta-analysis of eight RCTs employing thiazolidinediones in biopsy-proven NASH patients demonstrated that pioglitazone was associated with improvement in fibrosis of any stage (OR: 1.77 [1.15–2.72], p = 0.009), including advanced fibrosis (OR: 4.53 [1.52–13.52], p = 0.007) [165]. In addition, some preliminary results also suggested potential benefits of the sodium–glucose cotransporter 2 (SGLT 2) inhibitors on the markers of liver fibrosis [166,167].
Transaminases levels. Some studies have found that a higher ALT level or a low AST/ALT ratio are associated with significant fibrosis (or development of progressive fibrosis), but ALT is rather a minor independent factor that contributes to identifying fibrosis severity [14,49,168].
Drugs. Drug-induced steatosis/steatohepatitis is in fact an exclusion criterion for NAFLD, but patients with NAFLD and T2DM/obesity are often pluri-medicated, and there are some suggestions that certain drugs (i.e., acetaminophen, methotrexate, some antibiotics, steroids, tamoxifen, etc.) may elicit more severe (and perhaps more frequent) hepatotoxic effects and favor the progression to NASH/fibrosis in these patients [169,170,171]. The higher hepatotoxic susceptibility of NAFLD patients with diabetes and obesity is probably due to a certain metabolic and inflammatory environment, with hyperglycemia-induced oxidative stress, increased CYP2E1 expression and activity, lower ATP production, increased pro-inflammatory cytokines, mitochondrial dysfunction, etc. [169,172].
On the other hand, multiple drugs have been tested for their potential anti-inflammatory, cytoprotective, and antifibrotic effects in NAFLD patients [3,173,174]. Unfortunately, a great number of them have failed to demonstrate histologic improvement in fibrosis in clinical trials, but a few have shown some promising benefits [174,175,176]. However, there is no drug approved so far for NASH-associated fibrosis in patients with or without T2DM. Details regarding evidence from clinical trials are presented in Table 1 (only trials with positive results with regards to liver fibrosis or ongoing trials with fibrosis-specific endpoints are presented below) [158,177,178,179,180,181,182,183,184,185,186]. For a larger overview of drugs in development for NAFLD-related fibrosis, the reader is referred to references 174–176.

5. Liver Fibrogenesis and the Role of Metabolism

The progression of hepatic fibrosis is associated with chronic inflammation, loss of functional hepatocytes, and angiogenesis, but the key element is the over-production and accumulation of extracellular matrix (ECM), which is mainly produced by the activated HSCs that transdifferentiate into myofibroblasts [187,188]. The excessive ECM production seems to be accompanied by a change in its composition with advancing fibrosis, which might result in an increased resistance to degradation and a reduced resolution of fibrosis [187].
The excessive flux of FFAs and carbohydrates to the liver (resulting from exceeding dietary supply and dysfunctional adipose tissue) plays a prime role in the development of liver steatosis, lipotoxicity (generation of lipotoxic species), and subsequent pathogenetic mechanisms, while inflammation is the main driver of fibrogenesis, involving the immune system, vascular system, and soluble mediators [189]. Lipotoxicity results from excessive FFA influx and de novo lipogenesis and leads to mitochondrial dysfunction, oxidative and endoplasmic reticulum stress, apoptosis, and inflammation [173,187]. The hepatocyte lipoapoptosis is in fact a major driver of inflammation, and therefore, it is reasonable to assume that by reducing the metabolic injury, the inflammatory and fibrinogenic responses are attenuated [188]. Apart from the alterations in lipid metabolism, changes in glucose metabolism in the hepatocytes might apparently influence fibrogenesis through the crosstalk with the HSCs, mediated by proteins and perhaps other factors [190]. The hepatocyte stress and apoptosis stimulate inflammatory pathways with recruitment of macrophages and other immune cells, and further production of profibrogenic mediators, such as the TGFβ, which is a potent fibrogenic cytokine that activates HSCs [187]. Other profibrogenic mediators, like Hh ligands, damage-associated molecular patterns (DAMPs), osteopontin, etc., may also activate HSCs [187,188].
The processes of HSC activation and differentiation into myofibroblasts are accompanied by a significant metabolic reprogramming, needed to support these energy-requiring biological functions [187,188,190]. This consists of increased glycolysis, glutaminolysis, lipogenesis, and cholesterol accumulation in HSCs [190]. Some in vitro reports suggest that high concentrations of FFAs, cholesterol, and possibly also glucose may directly activate HSCs and induce the expression of fibrinogenic genes [191,192,193]. During HSC activation, there is a release of lipid droplets containing triglycerides and retinyl esters, an increase in FFAs β-oxidation, and a downregulation of transcription factors like sterol regulatory element-binding protein 1 (SREBP-1c) and PPAR-γ [194,195]. Thus, the pharmacological inhibition of lipogenesis, for example, by metabolic drugs (e.g., inhibitors of acetyl-CoA carboxylase (ACC) or lipoprotein lipase (LPL)) might attenuate fibrosis [190]. Insulin also seems to increase the expression of some fibrogenic signaling molecules, but the role of hyperglycemia and hyperinsulinemia in activating HSCs in the context of NAFLD-associated fibrosis is still not elucidated [162,194]. Hyperglycemia appears rather to amplify fibrogenesis induced by other factors [194]. The metabolic changes also affect the immune cells, mainly the macrophages, which have an important role during hepatic fibrogenesis [190]. Dysregulation of lipid metabolism and trafficking, with accumulation of toxic lipid metabolites, induce the pro-inflammatory phenotype of macrophages/Kupfer cells, while cholesterol crystals stimulate their activation and thus may contribute to fibrosis development [187,196,197,198].
To conclude, systemic and local metabolic derangements are involved in fibrogenic processes. During fibrogenesis, there is also an important metabolic reprogramming in HSCs, hepatocytes, and immune cells, engaging carbohydrate and lipid pathways to support the adaptation to high-energy demands. Preclinical data have indicated that the modulation of cell-intrinsic metabolism and reprogramming of the phenotype of these cells yields beneficial effects on liver fibrosis, but these results need to be substantiated by clinical studies [194].

6. Conclusions

Data in the literature point to the fact that between T2DM and NAFLD there is a bidirectional relationship, with one condition influencing the development and progression of the other. T2DM is associated with higher prevalence of NAFLD-associated fibrosis and favors its progression toward more advanced stages, while NAFLD increases the risk of diabetes, mainly in the presence of advanced fibrosis. There are a number of risk factors that impact the natural history of NAFLD, which has a dynamic and variable evolution. The timely recognition and control of the modifiable factors that determine a rapid progression of fibrosis are of critical importance, as the fibrosis stages are positively associated with the risk of liver-related and all-cause mortality.

Funding

This work was supported by the University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș Research Grant number 10126/5/17.12.2020. The research was carried out with the support of the Advanced Center for Medical and Pharmaceutical Research (CCAMF) of the George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Scientific Research Ethics Committee of the George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureş (number 1806 of 22.06.2022).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data is available upon request.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease. Diabetologia 2016, 59, 1121–1140. [Google Scholar] [CrossRef] [PubMed]
  2. Liebe, R.; Esposito, I.; Bock, H.H.; Vom Dahl, S.; Stindt, J.; Baumann, U.; Luedde, T.; Keitel, V. Diagnosis and management of secondary causes of steatohepatitis. J. Hepatol. 2021, 74, 1455–1471. [Google Scholar] [CrossRef] [PubMed]
  3. Cernea, S.; Raz, I. NAFLD in type 2 diabetes mellitus: Still many challenging questions. Diabetes Metab. Res. Rev. 2021, 37, e3386. [Google Scholar] [CrossRef] [PubMed]
  4. Targher, G.; Tilg, H.; Byrne, C.D. Non-alcoholic fatty liver disease: A multisystem disease requiring a multidisciplinary and holistic approach. Lancet Gastroenterol. Hepatol. 2021, 6, 578–588. [Google Scholar] [CrossRef]
  5. Fujii, H.; Kawada, N. Japan Study Group of Nafld Jsg-Nafld. The Role of Insulin Resistance and Diabetes in Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2020, 21, 3863. [Google Scholar] [CrossRef]
  6. Utzschneider, K.M.; Kahn, S.E. Review: The role of insulin resistance in nonalcoholic fatty liver disease. J. Clin. Endocrinol. Metab. 2006, 91, 4753–4761. [Google Scholar] [CrossRef]
  7. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef]
  8. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P.; et al. A multi-society Delphi consensus statement on new fatty liver disease nomenclature. J. Hepatol. 2023, 79, 1542–1556. [Google Scholar] [CrossRef]
  9. New NAFLD Nomenclature. Available online: https://www.aasld.org/new-nafld-nomenclature (accessed on 19 August 2023).
  10. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef]
  11. Younossi, Z.M.; Golabi, P.; de Avila, L.; Paik, J.M.; Srishord, M.; Fukui, N.; Qiu, Y.; Burns, L.; Afendy, A.; Nader, F. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J. Hepatol. 2019, 71, 793–801. [Google Scholar] [CrossRef]
  12. Henry, L.; Paik, J.; Younossi, Z.M. Review article: The epidemiologic burden of non-alcoholic fatty liver disease across the world. Aliment. Pharmacol. Ther. 2022, 56, 942–956. [Google Scholar] [CrossRef]
  13. Stefan, N.; Cusi, K. A global view of the interplay between non-alcoholic fatty liver disease and diabetes. Lancet Diabetes Endocrinol. 2022, 10, 284–296. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, H.W.; Kim, B.K.; Kim, S.U.; Park, J.Y.; Kim, D.Y.; Ahn, S.H.; Kim, K.J.; Han, K.H. Prevalence and Predictors of Significant Fibrosis Among Subjects with Transient Elastography-Defined Nonalcoholic Fatty Liver Disease. Dig. Dis. Sci. 2017, 62, 2150–2158. [Google Scholar] [CrossRef] [PubMed]
  15. Makker, J.; Tariq, H.; Kumar, K.; Ravi, M.; Shaikh, D.H.; Leung, V.; Hayat, U.; Hassan, M.T.; Patel, H.; Nayudu, S.; et al. Prevalence of advanced liver fibrosis and steatosis in type-2 diabetics with normal transaminases: A prospective cohort study. World J. Gastroenterol. 2021, 27, 523–533. [Google Scholar] [CrossRef] [PubMed]
  16. Ciardullo, S.; Monti, T.; Perseghin, G. High Prevalence of Advanced Liver Fibrosis Assessed by Transient Elastography Among U.S. Adults with Type 2 Diabetes. Diabetes Care 2021, 44, 519–525. [Google Scholar] [CrossRef] [PubMed]
  17. Cernea, S.; Onișor, D. Screening and interventions to prevent nonalcoholic fatty liver disease/nonalcoholic steatohepatitis-associated hepatocellular carcinoma. World J. Gastroenterol. 2023, 29, 286–309. [Google Scholar] [CrossRef] [PubMed]
  18. El-Serag, H.B.; Hampel, H.; Javadi, F. The association between diabetes and hepatocellular carcinoma: A systematic review of epidemiologic evidence. Clin. Gastroenterol. Hepatol. 2006, 4, 369–380. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, P.; Kang, D.; Cao, W.; Wang, Y.; Liu, Z. Diabetes mellitus and risk of hepatocellular carcinoma: A systematic review and meta-analysis. Diabetes Metab. Res. Rev. 2012, 28, 109–122. [Google Scholar] [CrossRef] [PubMed]
  20. Yang, J.D.; Ahmed, F.; Mara, K.C.; Addissie, B.D.; Allen, A.M.; Gores, G.J.; Roberts, L.R. Diabetes Is Associated with Increased Risk of Hepatocellular Carcinoma in Patients with Cirrhosis From Nonalcoholic Fatty Liver Disease. Hepatology 2020, 71, 907–916. [Google Scholar] [CrossRef]
  21. Huang, D.Q.; Noureddin, N.; Ajmera, V.; Amangurbanova, M.; Bettencourt, R.; Truong, E.; Gidener, T.; Siddiqi, H.; Majzoub, A.M.; Nayfeh, T.; et al. Type 2 diabetes, hepatic decompensation, and hepatocellular carcinoma in patients with non-alcoholic fatty liver disease: An individual participant-level data meta-analysis. Lancet Gastroenterol. Hepatol. 2023, 8, 829–836. [Google Scholar] [CrossRef]
  22. Aller de la Fuente, R.; Mora Cuadrado, N.; Tafur, C.; López Gómez, J.J.; Gómez de la Cuesta, S.; García Sánchez, M.C.; Antolin Melero, B.; de Luis Román, D.A. Histopathological differences in patients with biopsy-proven non-alcoholic fatty liver disease with and without type 2 diabetes. Endocrinol. Diabetes Nutr. 2018, 65, 354–360. [Google Scholar] [CrossRef]
  23. Puchakayala, B.K.; Verma, S.; Kanwar, P.; Hart, J.; Sanivarapu, R.R.; Mohanty, S.R. Histopathological differences utilizing the nonalcoholic fatty liver disease activity score criteria in diabetic (type 2 diabetes mellitus) and non-diabetic patients with nonalcoholic fatty liver disease. World J. Hepatol. 2015, 7, 2610–2618. [Google Scholar] [CrossRef] [PubMed]
  24. Bian, H.; Zhu, X.; Xia, M.; Yan, H.; Chang, X.; Hu, X.; Pan, B.; Guo, W.; Li, X.; Gao, X. Impact of TYPE 2 diabetes on nonalcoholic steatohepatitis and advanced fibrosis in patients with nonalcoholic fatty liver disease. Endocr. Pract. 2020, 26, 444–453. [Google Scholar] [CrossRef] [PubMed]
  25. Jarvis, H.; Craig, D.; Barker, R.; Spiers, G.; Stow, D.; Anstee, Q.M.; Hanratty, B. Metabolic risk factors and incident advanced liver disease in non-alcoholic fatty liver disease (NAFLD): A systematic review and meta-analysis of population-based observational studies. PLoS Med. 2020, 17, e1003100. [Google Scholar] [CrossRef] [PubMed]
  26. Kanwal, F.; Kramer, J.R.; Li, L.; Dai, J.; Natarajan, Y.; Yu, X.; Asch, S.M.; El-Serag, H.B. Effect of Metabolic Traits on the Risk of Cirrhosis and Hepatocellular Cancer in Nonalcoholic Fatty Liver Disease. Hepatology 2020, 71, 808–819. [Google Scholar] [CrossRef] [PubMed]
  27. Wild, S.H.; Morling, J.R.; McAllister, D.A.; Kerssens, J.; Fischbacher, C.; Parkes, J.; Roderick, P.J.; Sattar, N.; Byrne, C.D.; Scottish and Southampton Diabetes and Liver Disease Group; et al. Type 2 diabetes and risk of hospital admission or death for chronic liver diseases. J. Hepatol. 2016, 64, 1358–1364. [Google Scholar] [CrossRef]
  28. Liu, Z.J.; Yan, Y.J.; Weng, H.L.; Ding, H.G. Type 2 diabetes mellitus increases liver transplant-free mortality in patients with cirrhosis: A systematic review and meta-analysis. World J. Clin. Cases 2021, 9, 5514–5525. [Google Scholar] [CrossRef]
  29. Bertot, L.C.; Jeffrey, G.P.; de Boer, B.; MacQuillan, G.; Garas, G.; Chin, J.; Huang, Y.; Adams, L.A. Diabetes impacts prediction of cirrhosis and prognosis by non-invasive fibrosis models in non-alcoholic fatty liver disease. Liver Int. 2018, 38, 1793–1802. [Google Scholar] [CrossRef]
  30. Tada, T.; Toyoda, H.; Sone, Y.; Yasuda, S.; Miyake, N.; Kumada, T.; Tanaka, J. Type 2 diabetes mellitus: A risk factor for progression of liver fibrosis in middle-aged patients with non-alcoholic fatty liver disease. J. Gastroenterol. Hepatol. 2019, 34, 2011–2018. [Google Scholar] [CrossRef]
  31. Alexander, M.; Loomis, A.K.; van der Lei, J.; Duarte-Salles, T.; Prieto-Alhambra, D.; Ansell, D.; Pasqua, A.; Lapi, F.; Rijnbeek, P.; Mosseveld, M.; et al. Risks and clinical predictors of cirrhosis and hepatocellular carcinoma diagnoses in adults with diagnosed NAFLD: Real-world study of 18 million patients in four European cohorts. BMC Med. 2019, 17, 95. [Google Scholar] [CrossRef]
  32. Colosimo, S.; Tan, G.D.; Petroni, M.L.; Marchesini, G.; Tomlinson, J.W. Improved glycaemic control in patients with type 2 diabetes has a beneficial impact on NAFLD, independent of change in BMI or glucose lowering agent. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 640–648. [Google Scholar] [CrossRef] [PubMed]
  33. Ahlqvist, E.; Storm, P.; Käräjämäki, A.; Martinell, M.; Dorkhan, M.; Carlsson, A.; Vikman, P.; Prasad, R.B.; Aly, D.M.; Almgren, P.; et al. Novel subgroups of adult-onset diabetes and their association with outcomes: A data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018, 6, 361–369. [Google Scholar] [CrossRef] [PubMed]
  34. Zaharia, O.P.; Strassburger, K.; Strom, A.; Bönhof, G.J.; Karusheva, Y.; Antoniou, S.; Bódis, K.; Markgraf, D.F.; Burkart, V.; Müssig, K.; et al. Risk of diabetes-associated diseases in subgroups of patients with recent-onset diabetes: A 5-year follow-up study. Lancet Diabetes Endocrinol. 2019, 7, 684–694. [Google Scholar] [CrossRef] [PubMed]
  35. Zaharia, O.P.; Strassburger, K.; Knebel, B.; Kupriyanova, Y.; Karusheva, Y.; Wolkersdorfer, M.; Bódis, K.; Markgraf, D.F.; Burkart, V.; Hwang, J.H.; et al. Role of Patatin-Like Phospholipase Domain-Containing 3 Gene for Hepatic Lipid Content and Insulin Resistance in Diabetes. Diabetes Care. 2020, 43, 2161–2168. [Google Scholar] [CrossRef] [PubMed]
  36. Ballestri, S.; Zona, S.; Targher, G.; Romagnoli, D.; Baldelli, E.; Nascimbeni, F.; Roverato, A.; Guaraldi, G.; Lonardo, A. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J. Gastroenterol. Hepatol. 2016, 31, 936–944. [Google Scholar] [CrossRef] [PubMed]
  37. Morrison, A.E.; Zaccardi, F.; Khunti, K.; Davies, M.J. Causality between non-alcoholic fatty liver disease and risk of cardiovascular disease and type 2 diabetes: A meta-analysis with bias analysis. Liver Int. 2019, 39, 557–567. [Google Scholar] [CrossRef] [PubMed]
  38. Mantovani, A.; Petracca, G.; Beatrice, G.; Tilg, H.; Byrne, C.D.; 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. [Google Scholar] [CrossRef]
  39. Björkström, K.; Stål, P.; Hultcrantz, R.; Hagström, H. Histologic Scores for Fat and Fibrosis Associate with Development of Type 2 Diabetes in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2017, 15, 1461–1468. [Google Scholar] [CrossRef]
  40. Nasr, P.; Ignatova, S.; Kechagias, S.; Ekstedt, M. Natural history of nonalcoholic fatty liver disease: A prospective follow-up study with serial biopsies. Hepatol. Commun. 2017, 2, 199–210. [Google Scholar] [CrossRef]
  41. Ampuero, J.; Aller, R.; Gallego-Durán, R.; Crespo, J.; Calleja, J.L.; García-Monzón, C.; Gómez-Camarero, J.; Caballería, J.; Lo Iacono, O.; Ibañez, L.; et al. Significant fibrosis predicts new-onset diabetes mellitus and arterial hypertension in patients with NASH. J. Hepatol. 2020, 73, 17–25. [Google Scholar] [CrossRef]
  42. Yamazaki, H.; Tsuboya, T.; Tsuji, K.; Dohke, M.; Maguchi, H. Independent Association between Improvement of Nonalcoholic Fatty Liver Disease and Reduced Incidence of Type 2 Diabetes. Diabetes Care. 2015, 38, 1673–1679. [Google Scholar] [CrossRef] [PubMed]
  43. Cho, H.J.; Hwang, S.; Park, J.I.; Yang, M.J.; Hwang, J.C.; Yoo, B.M.; Lee, K.M.; Shin, S.J.; Lee, K.J.; Kim, J.H.; et al. Improvement of Nonalcoholic Fatty Liver Disease Reduces the Risk of Type 2 Diabetes Mellitus. Gut Liver. 2019, 13, 440–449. [Google Scholar] [CrossRef] [PubMed]
  44. Yamazaki, H.; Wang, J.; Tauchi, S.; Dohke, M.; Hanawa, N.; Katanuma, A.; Saisho, Y.; Kamitani, T.; Fukuhara, S.; Yamamoto, Y. Inverse Association between Fatty Liver at Baseline Ultrasonography and Remission of Type 2 Diabetes Over a 2-Year Follow-up Period. Clin. Gastroenterol. Hepatol. 2021, 19, 556–564.e5. [Google Scholar] [CrossRef]
  45. Xin, Z.; Huang, J.; Cao, Q.; Wang, J.; He, R.; Hou, T.; Ding, Y.; Lu, J.; Xu, M.; Wang, T.; et al. Nonalcoholic fatty liver disease in relation to the remission and progression along the glycemic continuum. J. Diabetes. 2022, 14, 606–619. [Google Scholar] [CrossRef] [PubMed]
  46. Henry, Z.H.; Argo, C.K. How to Identify the Patient with Nonalcoholic Steatohepatitis Who Will Progress to Cirrhosis. Gastroenterol. Clin. North. Am. 2020, 49, 45–62. [Google Scholar] [CrossRef]
  47. Calzadilla Bertot, L.; Adams, L.A. The Natural Course of Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2016, 17, 774. [Google Scholar] [CrossRef]
  48. Pais, R.; Maurel, T. Natural History of NAFLD. J. Clin. Med. 2021, 10, 1161. [Google Scholar] [CrossRef]
  49. Singh, S.; Allen, A.M.; Wang, Z.; Prokop, L.J.; Murad, M.H.; Loomba, R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: A systematic review and meta-analysis of paired-biopsy studies. Clin. Gastroenterol. Hepatol. 2015, 13, e1–e9. [Google Scholar] [CrossRef]
  50. Le, P.; Payne, J.Y.; Zhang, L.; Deshpande, A.; Rothberg, M.B.; Alkhouri, N.; Herman, W.; Hernandez, A.V.; Schleicher, M.; Ye, W.; et al. Disease State Transition Probabilities Across the Spectrum of NAFLD: A Systematic Review and Meta-Analysis of Paired Biopsy or Imaging Studies. Clin. Gastroenterol. Hepatol. 2023, 21, 1154–1168. [Google Scholar] [CrossRef]
  51. Ng, C.H.; Xiao, J.; Lim, W.H.; Chin, Y.H.; Yong, J.N.; Tan, D.J.H.; Tay, P.; Syn, N.; Foo, R.; Chan, M.; et al. Placebo effect on progression and regression in NASH: Evidence from a meta-analysis. Hepatology. 2022, 75, 1647–1661. [Google Scholar] [CrossRef]
  52. Pennisi, G.; Celsa, C.; Enea, M.; Vaccaro, M.; Di Marco, V.; Ciccioli, C.; Infantino, G.; La Mantia, C.; Parisi, S.; Vernuccio, F.; et al. Effect of pharmacological interventions and placebo on liver Histology in nonalcoholic steatohepatitis: A network meta-analysis. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 2279–2288. [Google Scholar] [CrossRef]
  53. Ampuero, J.; Gallego-Durán, R.; Maya-Miles, D.; Montero, R.; Gato, S.; Rojas, Á.; Gil, A.; Muñoz, R.; Romero-Gómez, M. Systematic review and meta-analysis: Analysis of variables influencing the interpretation of clinical trial results in NAFLD. J. Gastroenterol. 2022, 57, 357–371. [Google Scholar] [CrossRef] [PubMed]
  54. Giorda, C.; Forlani, G.; Manti, R.; Mazzella, N.; De Cosmo, S.; Rossi, M.C.; Nicolucci, A.; Russo, G.; Di Bartolo, P.; Ceriello, A.; et al. Occurrence over time and regression of nonalcoholic fatty liver disease in type 2 diabetes. Diabetes Metab. Res. Rev. 2017, 33, e2878. [Google Scholar] [CrossRef] [PubMed]
  55. Huang, D.Q.; Wilson, L.A.; Behling, C.; Kleiner, D.E.; Kowdley, K.V.; Dasarathy, S.; Amangurbanova, M.; Terrault, N.A.; Diehl, A.M.; Chalasani, N.; et al. Fibrosis Progression Rate in Biopsy-Proven Nonalcoholic Fatty Liver Disease Among People with Diabetes Versus People without Diabetes: A Multicenter Study. Gastroenterology. 2023, 165, 463–472.e5. [Google Scholar] [CrossRef] [PubMed]
  56. Nakahara, T.; Hyogo, H.; Yoneda, M.; Sumida, Y.; Eguchi, Y.; Fujii, H.; Ono, M.; Kawaguchi, T.; Imajo, K.; Aikata, H.; et al. Type 2 diabetes mellitus is associated with the fibrosis severity in patients with nonalcoholic fatty liver disease in a large retrospective cohort of Japanese patients. J. Gastroenterol. 2014, 49, 1477–1484. [Google Scholar] [CrossRef] [PubMed]
  57. Hagström, H.; Nasr, P.; Ekstedt, M.; Hammar, U.; Stål, P.; Hultcrantz, R.; Kechagias, S. Fibrosis stage but not NASH predicts mortality and time to development of severe liver disease in biopsy-proven NAFLD. J. Hepatol. 2017, 67, 1265–1273. [Google Scholar] [CrossRef] [PubMed]
  58. Dulai, P.S.; Singh, S.; Patel, J.; Soni, M.; Prokop, L.J.; Younossi, Z.; Sebastiani, G.; Ekstedt, M.; Hagstrom, H.; Nasr, P.; et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology 2017, 65, 1557–1565. [Google Scholar] [CrossRef] [PubMed]
  59. Cusi, K.; Isaacs, S.; Barb, D.; Basu, R.; Caprio, S.; Garvey, W.T.; Kashyap, S.; Mechanick, J.I.; Mouzaki, M.; Nadolsky, K.; et al. American Association of Clinical Endocrinology Clinical Practice Guideline for the Diagnosis and Management of Nonalcoholic Fatty Liver Disease in Primary Care and Endocrinology Clinical Settings: Co-Sponsored by the American Association for the Study of Liver Diseases (AASLD). Endocr. Pract. 2022, 28, 528–562. [Google Scholar] [PubMed]
  60. Jonas, W.; Schürmann, A. Genetic and epigenetic factors determining NAFLD risk. Mol. Metab. 2021, 50, 101111. [Google Scholar] [CrossRef]
  61. Valenti, L.; Al-Serri, A.; Daly, A.K.; Galmozzi, E.; Rametta, R.; Dongiovanni, P.; Nobili, V.; Mozzi, E.; Roviaro, G.; Vanni, E.; et al. Homozygosity for the patatin-like phospholipase-3/adiponutrin I148M polymorphism influences liver fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2010, 51, 1209–1217. [Google Scholar] [CrossRef]
  62. Sookoian, S.; Pirola, C.J. Meta-analysis of the influence of I148M variant of patatin-like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and histological severity of nonalcoholic fatty liver disease. Hepatology 2011, 53, 1883–1894. [Google Scholar] [CrossRef]
  63. Singal, A.G.; Manjunath, H.; Yopp, A.C.; Beg, M.S.; Marrero, J.A.; Gopal, P.; Waljee, A.K. The effect of PNPLA3 on fibrosis progression and development of hepatocellular carcinoma: A meta-analysis. Am. J. Gastroenterol. 2014, 109, 325–334. [Google Scholar] [CrossRef]
  64. Pingitore, P.; Dongiovanni, P.; Motta, B.M.; Meroni, M.; Lepore, S.M.; Mancina, R.M.; Pelusi, S.; Russo, C.; Caddeo, A.; Rossi, G.; et al. PNPLA3 overexpression results in reduction of proteins predisposing to fibrosis. Hum. Mol. Genet. 2016, 25, 5212–5222. [Google Scholar] [CrossRef]
  65. Vilar-Gomez, E.; Pirola, C.J.; Sookoian, S.; Wilson, L.A.; Belt, P.; Liang, T.; Liu, W.; Chalasani, N. Impact of the Association between PNPLA3 Genetic Variation and Dietary Intake on the Risk of Significant Fibrosis in Patients with NAFLD. Am. J. Gastroenterol. 2021, 116, 994–1006. [Google Scholar] [CrossRef]
  66. Chen, V.L.; Oliveri, A.; Miller, M.J.; Wijarnpreecha, K.; Du, X.; Chen, Y.; Cushing, K.C.; Lok, A.S.; Speliotes, E.K. PNPLA3 Genotype and Diabetes Identify Patients with Nonalcoholic Fatty Liver Disease at High Risk of Incident Cirrhosis. Gastroenterology 2023, 164, 966–977.e17. [Google Scholar] [CrossRef]
  67. Kozlitina, J.; Smagris, E.; Stender, S.; Nordestgaard, B.G.; Zhou, H.H.; Tybjærg-Hansen, A.; Vogt, T.F.; Hobbs, H.H.; Cohen, J.C. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 2014, 46, 352–356. [Google Scholar] [CrossRef]
  68. Liu, Y.L.; Reeves, H.L.; Burt, A.D.; Tiniakos, D.; McPherson, S.; Leathart, J.B.; Allison, M.E.; Alexander, G.J.; Piguet, A.C.; Anty, R.; et al. TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Nat. Commun. 2014, 5, 4309. [Google Scholar] [CrossRef] [PubMed]
  69. Dongiovanni, P.; Petta, S.; Maglio, C.; Fracanzani, A.L.; Pipitone, R.; Mozzi, E.; Motta, B.M.; Kaminska, D.; Rametta, R.; Grimaudo, S.; et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology 2015, 61, 506–514. [Google Scholar] [CrossRef] [PubMed]
  70. Krawczyk, M.; Rau, M.; Schattenberg, J.M.; Bantel, H.; Pathil, A.; Demir, M.; Kluwe, J.; Boettler, T.; Lammert, F.; Geier, A.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  71. Teo, K.; Abeysekera, K.W.M.; Adams, L.; Aigner, E.; Anstee, Q.M.; Banales, J.M.; Banerjee, R.; Basu, P.; Berg, T.; Bhatnagar, P.; et al. rs641738C>T near MBOAT7 is associated with liver fat, ALT and fibrosis in NAFLD: A meta-analysis. J. Hepatol. 2021, 74, 20–30. [Google Scholar] [CrossRef] [PubMed]
  72. Del Campo, J.A.; Gallego-Durán, R.; Gallego, P.; Grande, L. Genetic and Epigenetic Regulation in Nonalcoholic Fatty Liver Disease (NAFLD). Int. J. Mol. Sci. 2018, 19, 911. [Google Scholar] [CrossRef]
  73. Petta, S.; Miele, L.; Bugianesi, E.; Cammà, C.; Rosso, C.; Boccia, S.; Cabibi, D.; Di Marco, V.; Grimaudo, S.; Grieco, A.; et al. Glucokinase regulatory protein gene polymorphism affects liver fibrosis in non-alcoholic fatty liver disease. PLoS ONE 2014, 9, e87523, Erratum in PLoS ONE 2014, 9, e92497. [Google Scholar] [CrossRef]
  74. Eslam, M.; Valenti, L.; Romeo, S. Genetics and epigenetics of NAFLD and NASH: Clinical impact. J. Hepatol. 2018, 68, 268–279. [Google Scholar] [CrossRef]
  75. Govaere, O.; Cockell, S.; Tiniakos, D.; Queen, R.; Younes, R.; Vacca, M.; Alexander, L.; Ravaioli, F.; Palmer, J.; Petta, S.; et al. Transcriptomic profiling across the nonalcoholic fatty liver disease spectrum reveals gene signatures for steatohepatitis and fibrosis. Sci. Transl. Med. 2020, 12, eaba4448. [Google Scholar] [CrossRef]
  76. Villani, R.; Magnati, G.P.; De Girolamo, G.; Sangineto, M.; Romano, A.D.; Cassano, T.; Serviddio, G. Genetic Polymorphisms and Clinical Features in Diabetic Patients with Fatty Liver: Results From a Single-Center Experience in Southern Italy. Front. Med. 2021, 8, 737759. [Google Scholar] [CrossRef]
  77. Lavrado, N.C.; Salles, G.F.; Cardoso, C.R.L.; de França, P.H.C.; Melo, M.F.D.G.G.; Leite, N.C.; Villela-Nogueira, C.A. Impact of PNPLA3 and TM6SF2 polymorphisms on the prognosis of patients with MASLD and type 2 diabetes mellitus. Liver Int. 2024; Online ahead of print. [Google Scholar] [CrossRef]
  78. Gellert-Kristensen, H.; Richardson, T.G.; Davey Smith, G.; Nordestgaard, B.G.; Tybjaerg-Hansen, A.; Stender, S. Combined Effect of PNPLA3, TM6SF2, and HSD17B13 Variants on Risk of Cirrhosis and Hepatocellular Carcinoma in the General Population. Hepatology 2020, 72, 845–856. [Google Scholar] [CrossRef] [PubMed]
  79. Choudhary, N.S.; Duseja, A. Genetic and epigenetic disease modifiers: Non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). Transl. Gastroenterol. Hepatol. 2021, 6, 2. [Google Scholar] [CrossRef]
  80. Zeybel, M.; Hardy, T.; Robinson, S.M.; Fox, C.; Anstee, Q.M.; Ness, T.; Masson, S.; Mathers, J.C.; French, J.; White, S.; et al. Differential DNA methylation of genes involved in fibrosis progression in non-alcoholic fatty liver disease and alcoholic liver disease. Clin. Epigenetics. 2015, 7, 25. [Google Scholar] [CrossRef] [PubMed]
  81. Hardy, T.; Zeybel, M.; Day, C.P.; Dipper, C.; Masson, S.; McPherson, S.; Henderson, E.; Tiniakos, D.; White, S.; French, J.; et al. Plasma DNA methylation: A potential biomarker for stratification of liver fibrosis in non-alcoholic fatty liver disease. Gut 2017, 66, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  82. Murphy, S.K.; Yang, H.; Moylan, C.A.; Pang, H.; Dellinger, A.; Abdelmalek, M.F.; Garrett, M.E.; Ashley-Koch, A.; Suzuki, A.; Tillmann, H.L.; et al. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 1076–1087. [Google Scholar] [CrossRef]
  83. Cheung, O.; Puri, P.; Eicken, C.; Contos, M.J.; Mirshahi, F.; Maher, J.W.; Kellum, J.M.; Min, H.; Luketic, V.A.; Sanyal, A.J. Nonalcoholic steatohepatitis is associated with altered hepatic MicroRNA expression. Hepatology 2008, 48, 1810–1820. [Google Scholar] [CrossRef]
  84. Zarrinpar, A.; Gupta, S.; Maurya, M.R.; Subramaniam, S.; Loomba, R. Serum microRNAs explain discordance of non-alcoholic fatty liver disease in monozygotic and dizygotic twins: A prospective study. Gut 2016, 65, 1546–1554. [Google Scholar] [CrossRef]
  85. Hanson, A.; Wilhelmsen, D.; DiStefano, J.K. The Role of Long Non-Coding RNAs (lncRNAs) in the Development and Progression of Fibrosis Associated with Nonalcoholic Fatty Liver Disease (NAFLD). Noncoding RNA 2018, 4, 18. [Google Scholar] [CrossRef]
  86. Riazi, K.; Swain, M.G.; Congly, S.E.; Kaplan, G.G.; Shaheen, A.A. Race and Ethnicity in Non-Alcoholic Fatty Liver Disease (NAFLD): A Narrative Review. Nutrients 2022, 14, 4556. [Google Scholar] [CrossRef]
  87. Rich, N.E.; Oji, S.; Mufti, A.R.; Browning, J.D.; Parikh, N.D.; Odewole, M.; Mayo, H.; Singal, A.G. Racial and Ethnic Disparities in Nonalcoholic Fatty Liver Disease Prevalence, Severity, and Outcomes in the United States: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2018, 16, 198–210.e2. [Google Scholar] [CrossRef] [PubMed]
  88. Truong, E.; Yeo, Y.H.; Cook-Wiens, G.; Muthiah, M.; Yang, J.D.; Sundaram, V.; Chang, D.; Todo, T.; Kim, I.K.; Lu, S.C.; et al. Nonalcoholic fatty liver disease prevalence and severity in Asian Americans from the national health and nutrition examination surveys 2017–2018. Hepatol Commun. 2022, 6, 2253–2261. [Google Scholar] [CrossRef] [PubMed]
  89. Alqahtani, S.A.; Schattenberg, J.M. NAFLD in the Elderly. Clin. Interv. Aging. 2021, 16, 1633–1649. [Google Scholar] [CrossRef] [PubMed]
  90. Miyaaki, H.; Ichikawa, T.; Nakao, K.; Yatsuhashi, H.; Furukawa, R.; Ohba, K.; Omagari, K.; Kusumoto, Y.; Yanagi, K.; Inoue, O.; et al. Clinicopathological study of nonalcoholic fatty liver disease in Japan: The risk factors for fibrosis. Liver Int. 2008, 28, 519–524. [Google Scholar] [CrossRef] [PubMed]
  91. Frith, J.; Day, C.P.; Henderson, E.; Burt, A.D.; Newton, J.L. Non-alcoholic fatty liver disease in older people. Gerontology 2009, 55, 607–613. [Google Scholar] [CrossRef] [PubMed]
  92. Nagral, A.; Bangar, M.; Menezes, S.; Bhatia, S.; Butt, N.; Ghosh, J.; Manchanayake, J.H.; Mahtab, M.A.; Singh, S.P. Gender Differences in Nonalcoholic Fatty Liver Disease. Euroasian J. Hepatogastroenterol. 2022, 12 (Suppl. S1), S19–S25. [Google Scholar] [PubMed]
  93. Balakrishnan, M.; Patel, P.; Dunn-Valadez, S.; Dao, C.; Khan, V.; Ali, H.; El-Serag, L.; Hernaez, R.; Sisson, A.; Thrift, A.P.; et al. Women Have a Lower Risk of Nonalcoholic Fatty Liver Disease but a Higher Risk of Progression vs Men: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2021, 19, 61–71.e15. [Google Scholar] [CrossRef]
  94. Sarkar, M.A.; Suzuki, A.; Abdelmalek, M.F.; Yates, K.P.; Wilson, L.A.; Bass, N.M.; Gill, R.; Cedars, M.; Terrault, N.; NASH Clinical Research Network. Testosterone is Associated with Nonalcoholic Steatohepatitis and Fibrosis in Premenopausal Women with NAFLD. Clin. Gastroenterol. Hepatol. 2021, 19, 1267–1274.e1. [Google Scholar] [CrossRef]
  95. Zhang, Z.; Chen, C.; Wang, Y.; Wang, N.; Chen, Y.; Lu, Y.; Xia, F. The associations of total testosterone with probable nonalcoholic steatohepatitis and nonalcoholic fatty liver disease fibrotic progression in men with type 2 diabetes: A cross-sectional study. Eur. J. Med. Res. 2022, 27, 307. [Google Scholar] [CrossRef]
  96. Hazlehurst, J.M.; Tomlinson, J.W. Non-alcoholic fatty liver disease in common endocrine disorders. Eur. J. Endocrinol. 2013, 169, R27–R37. [Google Scholar] [CrossRef] [PubMed]
  97. Arefhosseini, S.; Ebrahimi-Mameghani, M.; Najafipour, F.; Tutunchi, H. Non-alcoholic fatty liver disease across endocrinopathies: Interaction with sex hormones. Front. Endocrinol. 2022, 13, 1032361. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, Y.; Li, J.; Liu, H. Correlation between the thyroid hormone levels and nonalcoholic fatty liver disease in type 2 diabetic patients with normal thyroid function. BMC Endocr. Disord. 2022, 22, 144. [Google Scholar] [CrossRef] [PubMed]
  99. Tsompanaki, E.; Thanapirom, K.; Papatheodoridi, M.; Parikh, P.; Chotai de Lima, Y.; Tsochatzis, E.A. Systematic Review and Meta-analysis: The Role of Diet in the Development of Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2023, 21, 1462–1474.e24. [Google Scholar] [CrossRef] [PubMed]
  100. Vilar-Gomez, E.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Friedman, S.L.; Diago, M.; Romero-Gomez, M. Weight Loss Through Lifestyle Modification Significantly Reduces Feature of Nonalcoholic Steatohepatitis. Gastroenterology 2015, 149, 367–378.e5. [Google Scholar] [CrossRef] [PubMed]
  101. Miryan, M.; Darbandi, M.; Moradi, M.; Najafi, F.; Soleimani, D.; Pasdar, Y. Relationship between the Mediterranean diet and risk of hepatic fibrosis in patients with non-alcoholic fatty liver disease: A cross-sectional analysis of the RaNCD cohort. Front. Nutr. 2023, 10, 1062008. [Google Scholar] [CrossRef] [PubMed]
  102. Perez-Diaz-Del-Campo, N.; Castelnuovo, G.; Rosso, C.; Nicolosi, A.; Guariglia, M.; Dileo, E.; Armandi, A.; Caviglia, G.P.; Bugianesi, E. Impact of Health Related QoL and Mediterranean Diet on Liver Fibrosis in Patients with NAFLD. Nutrients 2023, 15, 3018. [Google Scholar] [CrossRef] [PubMed]
  103. Kouvari, M.; Boutari, C.; Chrysohoou, C.; Fragkopoulou, E.; Antonopoulou, S.; Tousoulis, D.; Pitsavos, C.; Panagiotakos, D.B.; Mantzoros, C.S.; ATTICA study Investigators. Mediterranean diet is inversely associated with steatosis and fibrosis and decreases ten-year diabetes and cardiovascular risk in NAFLD subjects: Results from the ATTICA prospective cohort study. Clin. Nutr. 2021, 40, 3314–3324. [Google Scholar] [CrossRef] [PubMed]
  104. Baratta, F.; Cammisotto, V.; Tozzi, G.; Coronati, M.; Bartimoccia, S.; Castellani, V.; Nocella, C.; D’Amico, A.; Angelico, F.; Carnevale, R.; et al. High Compliance to Mediterranean Diet Associates with Lower Platelet Activation and Liver Collagen Deposition in Patients with Nonalcoholic Fatty Liver Disease. Nutrients 2022, 14, 1209. [Google Scholar] [CrossRef] [PubMed]
  105. Curci, R.; Bianco, A.; Franco, I.; Bonfiglio, C.; Campanella, A.; Mirizzi, A.; Giannuzzi, V.; Cozzolongo, R.; Veronese, N.; Osella, A.R. Lifestyle Modification: Evaluation of the Effects of Physical Activity and Low-Glycemic-Index Mediterranean Diet on Fibrosis Score. Nutrients 2023, 15, 3520. [Google Scholar] [CrossRef] [PubMed]
  106. Katsagoni, C.N.; Papatheodoridis, G.V.; Ioannidou, P.; Deutsch, M.; Alexopoulou, A.; Papadopoulos, N.; Papageorgiou, M.V.; Fragopoulou, E.; Kontogianni, M.D. Improvements in clinical characteristics of patients with non-alcoholic fatty liver disease, after an intervention based on the Mediterranean lifestyle: A randomised controlled clinical trial. Br. J. Nutr. 2018, 120, 164–175. [Google Scholar] [CrossRef] [PubMed]
  107. Soleimani, D.; Ranjbar, G.; Rezvani, R.; Goshayeshi, L.; Razmpour, F.; Nematy, M. Dietary patterns in relation to hepatic fibrosis among patients with nonalcoholic fatty liver disease. Diabetes Metab. Syndr. Obes. 2019, 12, 315–324. [Google Scholar] [CrossRef] [PubMed]
  108. Gao, V.; Long, M.T.; Singh, S.R.; Kim, Y.; Zhang, X.; Rogers, G.; Jacques, P.F.; Levy, D.; Ma, J. A Healthy Diet is Associated with a Lower Risk of Hepatic Fibrosis. J. Nutr. 2023, 153, 1587–1596. [Google Scholar] [CrossRef] [PubMed]
  109. Johari, M.I.; Yusoff, K.; Haron, J.; Nadarajan, C.; Ibrahim, K.N.; Wong, M.S.; Hafidz, M.I.A.; Chua, B.E.; Hamid, N.; Arifin, W.N.; et al. A Randomised Controlled Trial on the Effectiveness and Adherence of Modified Alternate-day Calorie Restriction in Improving Activity of Non-Alcoholic Fatty Liver Disease. Sci. Rep. 2019, 9, 11232. [Google Scholar] [CrossRef]
  110. López-Bautista, F.; Barbero-Becerra, V.J.; Ríos, M.Y.; Ramírez-Cisneros, M.Á.; Sánchez-Pérez, C.A.; Ramos-Ostos, M.H.; Uribe, M.; Chávez-Tapia, N.C.; Juárez-Hernández, E. Dietary consumption and serum pattern of bioactive fatty acids in NAFLD patients. Ann. Hepatol. 2020, 19, 482–488. [Google Scholar] [CrossRef]
  111. Guerrerio, A.L.; Colvin, R.M.; Schwartz, A.K.; Molleston, J.P.; Murray, K.F.; Diehl, A.; Mohan, P.; Schwimmer, J.B.; Lavine, J.E.; Torbenson, M.S.; et al. Choline intake in a large cohort of patients with nonalcoholic fatty liver disease. Am. J. Clin. Nutr. 2012, 95, 892–900. [Google Scholar] [CrossRef]
  112. Ebadi, M.; Ip, S.; Bhanji, R.A.; Montano-Loza, A.J. Effect of Coffee Consumption on Non-Alcoholic Fatty Liver Disease Incidence, Prevalence and Risk of Significant Liver Fibrosis: Systematic Review with Meta-Analysis of Observational Studies. Nutrients 2021, 13, 3042. [Google Scholar] [CrossRef]
  113. Khodami, B.; Hatami, B.; Yari, Z.; Alavian, S.M.; Sadeghi, A.; Varkaneh, H.K.; Santos, H.O.; Hekmatdoost, A. Effects of a low free sugar diet on the management of nonalcoholic fatty liver disease: A randomized clinical trial. Eur. J. Clin. Nutr. 2022, 76, 987–994. [Google Scholar] [CrossRef]
  114. Protopapas, A.A.; Cholongitas, E.; Chrysavgis, L.; Tziomalos, K. Alcohol consumption in patients with nonalcoholic fatty liver disease: Yes, or no? Ann. Gastroenterol. 2021, 34, 476–486. [Google Scholar] [CrossRef]
  115. Chang, Y.; Cho, Y.K.; Kim, Y.; Sung, E.; Ahn, J.; Jung, H.S.; Yun, K.E.; Shin, H.; Ryu, S. Nonheavy Drinking and Worsening of Noninvasive Fibrosis Markers in Nonalcoholic Fatty Liver Disease: A Cohort Study. Hepatology 2019, 69, 64–75. [Google Scholar] [CrossRef]
  116. Ekstedt, M.; Franzén, L.E.; Holmqvist, M.; Bendtsen, P.; Mathiesen, U.L.; Bodemar, G.; Kechagias, S. Alcohol consumption is associated with progression of hepatic fibrosis in non-alcoholic fatty liver disease. Scand. J. Gastroenterol. 2009, 44, 366–374. [Google Scholar] [CrossRef]
  117. Åberg, F.; Puukka, P.; Salomaa, V.; Männistö, S.; Lundqvist, A.; Valsta, L.; Perola, M.; Färkkilä, M.; Jula, A. Risks of Light and Moderate Alcohol Use in Fatty Liver Disease: Follow-Up of Population Cohorts. Hepatology 2020, 71, 835–848. [Google Scholar] [CrossRef]
  118. Koo, B.K.; Kim, D.; Joo, S.K.; Kim, J.H.; Chang, M.S.; Kim, B.G.; Lee, K.L.; Kim, W. Sarcopenia is an independent risk factor for non-alcoholic steatohepatitis and significant fibrosis. J. Hepatol. 2017, 66, 123–131. [Google Scholar] [CrossRef] [PubMed]
  119. Petta, S.; Ciminnisi, S.; Di Marco, V.; Cabibi, D.; Cammà, C.; Licata, A.; Marchesini, G.; Craxì, A. Sarcopenia is associated with severe liver fibrosis in patients with non-alcoholic fatty liver disease. Aliment. Pharmacol. Ther. 2017, 45, 510–518. [Google Scholar] [CrossRef] [PubMed]
  120. Hsieh, Y.C.; Joo, S.K.; Koo, B.K.; Lin, H.C.; Kim, W. Muscle alterations are independently associated with significant fibrosis in patients with nonalcoholic fatty liver disease. Liver Int. 2021, 41, 494–504. [Google Scholar] [CrossRef]
  121. Lee, Y.H.; Kim, S.U.; Song, K.; Park, J.Y.; Kim, D.Y.; Ahn, S.H.; Lee, B.W.; Kang, E.S.; Cha, B.S.; Han, K.H. Sarcopenia is associated with significant liver fibrosis independently of obesity and insulin resistance in nonalcoholic fatty liver disease: Nationwide surveys (KNHANES 2008–2011). Hepatology 2016, 63, 776–786. [Google Scholar] [CrossRef] [PubMed]
  122. Shida, T.; Oshida, N.; Oh, S.; Okada, K.; Shoda, J. Progressive reduction in skeletal muscle mass to visceral fat area ratio is associated with a worsening of the hepatic conditions of non-alcoholic fatty liver disease. Diabetes Metab. Syndr. Obes. 2019, 12, 495–503. [Google Scholar] [CrossRef]
  123. Hsieh, Y.C.; Joo, S.K.; Koo, B.K.; Lin, H.C.; Lee, D.H.; Chang, M.S.; Park, J.H.; So, Y.H.; Kim, W.; Innovative Target Exploration of NAFLD (ITEN) Consortium. Myosteatosis, but not Sarcopenia, Predisposes NAFLD Subjects to Early Steatohepatitis and Fibrosis Progression. Clin. Gastroenterol. Hepatol. 2023, 21, 388–397.e10. [Google Scholar] [CrossRef]
  124. Armandi, A.; Rosso, C.; Nicolosi, A.; Caviglia, G.P.; Abate, M.L.; Olivero, A.; D’Amato, D.; Vernero, M.; Gaggini, M.; Saracco, G.M.; et al. Crosstalk between Irisin Levels, Liver Fibrogenesis and Liver Damage in Non-Obese, Non-Diabetic Individuals with Non-Alcoholic Fatty Liver Disease. J. Clin. Med. 2022, 11, 635. [Google Scholar] [CrossRef]
  125. Kuchay, M.S.; Martínez-Montoro, J.I.; Kaur, P.; Fernández-García, J.C.; Ramos-Molina, B. Non-alcoholic fatty liver disease-related fibrosis and sarcopenia: An altered liver-muscle crosstalk leading to increased mortality risk. Ageing Res. Rev. 2022, 80, 101696. [Google Scholar] [CrossRef]
  126. Zhu, W.; Sahar, N.E.; Javaid, H.M.A.; Pak, E.S.; Liang, G.; Wang, Y.; Ha, H.; Huh, J.Y. Exercise-Induced Irisin Decreases Inflammation and Improves NAFLD by Competitive Binding with MD2. Cells 2021, 10, 3306. [Google Scholar] [CrossRef]
  127. Zhao, J.; Qiao, L.; Dong, J.; Wu, R. Antioxidant Effects of Irisin in Liver Diseases: Mechanistic Insights. Oxid. Med. Cell Longev. 2022, 2022, 3563518. [Google Scholar] [CrossRef]
  128. Oh, S.; So, R.; Shida, T.; Matsuo, T.; Kim, B.; Akiyama, K.; Isobe, T.; Okamoto, Y.; Tanaka, K.; Shoda, J. High-Intensity Aerobic Exercise Improves Both Hepatic Fat Content and Stiffness in Sedentary Obese Men with Nonalcoholic Fatty Liver Disease. Sci. Rep. 2017, 7, 43029. [Google Scholar] [CrossRef] [PubMed]
  129. O’Gorman, P.; Naimimohasses, S.; Monaghan, A.; Kennedy, M.; Melo, A.M.; Ní Fhloinn, D.; Doherty, D.G.; Beddy, P.; Finn, S.P.; Moore, J.B.; et al. Improvement in histological endpoints of MAFLD following a 12-week aerobic exercise intervention. Aliment. Pharmacol. Ther. 2020, 52, 1387–1398. [Google Scholar] [CrossRef]
  130. Chen, G.; Banini, B.; Do, A.; Lim, J.K. The independent effect of exercise on biopsy-proven non-alcoholic fatty liver disease: A systematic review. Clin. Mol. Hepatol. 2023, 29, 414–416. [Google Scholar] [CrossRef]
  131. Vallianou, N.; Christodoulatos, G.S.; Karampela, I.; Tsilingiris, D.; Magkos, F.; Stratigou, T.; Kounatidis, D.; Dalamaga, M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Non-Alcoholic Fatty Liver Disease: Current Evidence and Perspectives. Biomolecules 2021, 12, 56. [Google Scholar] [CrossRef]
  132. Chen, D.; Le, T.H.; Shahidipour, H.; Read, S.A.; Ahlenstiel, G. The Role of Gut-Derived Microbial Antigens on Liver Fibrosis Initiation and Progression. Cells 2019, 8, 1324. [Google Scholar] [CrossRef]
  133. Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The role of the gut microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef]
  134. de Faria Ghetti, F.; Oliveira, D.G.; de Oliveira, J.M.; de Castro Ferreira, L.E.V.V.; Cesar, D.E.; Moreira, A.P.B. Influence of gut microbiota on the development and progression of nonalcoholic steatohepatitis. Eur. J. Nutr. 2018, 57, 861–876. [Google Scholar] [CrossRef]
  135. Bastian, W.P.; Hasan, I.; Lesmana, C.R.A.; Rinaldi, I.; Gani, R.A. Gut Microbiota Profiles in Nonalcoholic Fatty Liver Disease and Its Possible Impact on Disease Progression Evaluated with Transient Elastography: Lesson Learnt from 60 Cases. Case Rep. Gastroenterol. 2019, 13, 125–133. [Google Scholar] [CrossRef]
  136. Loomba, R.; Seguritan, V.; Li, W.; Long, T.; Klitgord, N.; Bhatt, A.; Dulai, P.S.; Caussy, C.; Bettencourt, R.; Highlander, S.K.; et al. Gut Microbiome-Based Metagenomic Signature for Non-invasive Detection of Advanced Fibrosis in Human Nonalcoholic Fatty Liver Disease. Cell Metab. 2017, 25, 1054–1062.e5. [Google Scholar] [CrossRef]
  137. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016, 63, 764–775. [Google Scholar] [CrossRef] [PubMed]
  138. Gómez-Pérez, A.M.; Ruiz-Limón, P.; Salas-Salvadó, J.; Vioque, J.; Corella, D.; Fitó, M.; Vidal, J.; Atzeni, A.; Torres-Collado, L.; Álvarez-Sala, A.; et al. Gut microbiota in nonalcoholic fatty liver disease: A PREDIMED-Plus trial sub analysis. Gut Microbes. 2023, 15, 2223339. [Google Scholar] [CrossRef] [PubMed]
  139. Man, S.; Lv, J.; Yu, C.; Deng, Y.; Yin, J.; Wang, B.; Li, L.; Liu, H. Association between metabolically healthy obesity and non-alcoholic fatty liver disease. Hepatol. Int. 2022, 16, 1412–1423. [Google Scholar] [CrossRef] [PubMed]
  140. Saponaro, C.; Sabatini, S.; Gaggini, M.; Carli, F.; Rosso, C.; Positano, V.; Armandi, A.; Caviglia, G.P.; Faletti, R.; Bugianesi, E.; et al. Adipose tissue dysfunction and visceral fat are associated with hepatic insulin resistance and severity of NASH even in lean individuals. Liver Int. 2022, 42, 2418–2427. [Google Scholar] [CrossRef] [PubMed]
  141. Hernández-Conde, M.; Llop, E.; Carrillo, C.F.; Tormo, B.; Abad, J.; Rodriguez, L.; Perelló, C.; Gomez, M.L.; Martínez-Porras, J.L.; Puga, N.F.; et al. Estimation of visceral fat is useful for the diagnosis of significant fibrosis in patients with non-alcoholic fatty liver disease. World J. Gastroenterol. 2020, 26, 6658–6668. [Google Scholar] [CrossRef] [PubMed]
  142. Kim, Y.; Chang, Y.; Cho, Y.K.; Ahn, J.; Shin, H.; Ryu, S. Metabolically healthy versus unhealthy obesity and risk of fibrosis progression in non-alcoholic fatty liver disease. Liver Int. 2019, 39, 1884–1894. [Google Scholar] [CrossRef] [PubMed]
  143. Kim, Y.; Chang, Y.; Cho, Y.K.; Ahn, J.; Shin, H.; Ryu, S. Obesity and Weight Gain Are Associated with Progression of Fibrosis in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2019, 17, 543–550.e2. [Google Scholar] [CrossRef] [PubMed]
  144. Julián, M.T.; Ballesta, S.; Pera, G.; Pérez-Montes de Oca, A.; Soldevila, B.; Caballería, L.; Morillas, R.; Expósito, C.; Martínez-Escudé, A.; Puig-Domingo, M.; et al. Abdominal obesity and dsyglycemia are risk factors for liver fibrosis progression in NAFLD subjects: A population-based study. Front. Endocrinol. 2023, 13, 1051958. [Google Scholar] [CrossRef]
  145. Koutoukidis, D.A.; Jebb, S.A.; Tomlinson, J.W.; Cobbold, J.F.; Aveyard, P. Association of Weight Changes with Changes in Histological Features and Blood Markers in Nonalcoholic Steatohepatitis. Clin. Gastroenterol. Hepatol. 2022, 20, e538–e547. [Google Scholar] [CrossRef] [PubMed]
  146. Pais, R.; Charlotte, F.; Fedchuk, L.; Bedossa, P.; Lebray, P.; Poynard, T.; Ratziu, V.; LIDO Study Group. A systematic review of follow-up biopsies reveals disease progression in patients with non-alcoholic fatty liver. J. Hepatol. 2013, 59, 550–556. [Google Scholar] [CrossRef]
  147. McPherson, S.; Hardy, T.; Henderson, E.; Burt, A.D.; Day, C.P.; Anstee, Q.M. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: Implications for prognosis and clinical management. J. Hepatol. 2015, 62, 1148–1155. [Google Scholar] [CrossRef]
  148. Adams, L.A.; Sanderson, S.; Lindor, K.D.; Angulo, P. The histological course of nonalcoholic fatty liver disease: A longitudinal study of 103 patients with sequential liver biopsies. J. Hepatol. 2005, 42, 132–138. [Google Scholar] [CrossRef]
  149. Wajsbrot, N.B.; Leite, N.C.; Franca, P.H.C.; Cardoso, C.R.L.; Salles, G.F.; Villela-Nogueira, C.A. Parental History of Type 2 Diabetes Mellitus and PNPLA3 Polymorphism Increase the Risk of Severe Stages of Nonalcoholic Fatty Liver Disease. Dig. Dis. Sci. 2023, 69, 634–642. [Google Scholar] [CrossRef] [PubMed]
  150. Abbasi, A.; Corpeleijn, E.; van der Schouw, Y.T.; Stolk, R.P.; Spijkerman, A.; van der, A.D.L.; Navis, G.; Bakker, S.J.; Beulens, J.W. Parental history of type 2 diabetes and cardiometabolic biomarkers in offspring. Eur. J. Clin. Investig. 2012, 42, 974–982. [Google Scholar] [CrossRef]
  151. De Pergola, G.; Castellana, F.; Zupo, R.; De Nucci, S.; Panza, F.; Castellana, M.; Lampignano, L.; Di Chito, M.; Triggiani, V.; Sardone, R.; et al. A family history of type 2 diabetes as a predictor of fatty liver disease in diabetes-free individuals with excessive body weight. Sci. Rep. 2021, 11, 24084. [Google Scholar] [CrossRef]
  152. Schiaffini, R.; Liccardo, D.; Alisi, A.; Benevento, D.; Cappa, M.; Cianfarani, S.; Nobili, V. Early Glucose Derangement Detected by Continuous Glucose Monitoring and Progression of Liver Fibrosis in Nonalcoholic Fatty Liver Disease: An Independent Predictive Factor? Horm. Res. Paediatr. 2016, 85, 29–34. [Google Scholar] [CrossRef]
  153. Hashiba, M.; Ono, M.; Hyogo, H.; Ikeda, Y.; Masuda, K.; Yoshioka, R.; Ishikawa, Y.; Nagata, Y.; Munekage, K.; Ochi, T.; et al. Glycemic variability is an independent predictive factor for development of hepatic fibrosis in nonalcoholic fatty liver disease. PLoS ONE 2013, 8, e76161. [Google Scholar] [CrossRef]
  154. Chang, X.; Bian, H.; Xia, M.; Zhu, X.; Sun, X.; Yang, X.; Gao, J.; Lin, H.; Yan, H.; Gao, X. Postprandial glucose is correlated with an increasing risk of liver fibrosis in Chinese patients with nonalcoholic fatty liver disease. Diabetes Metab. 2022, 48, 101377. [Google Scholar] [CrossRef] [PubMed]
  155. Sako, S.; Takeshita, Y.; Takayama, H.; Goto, H.; Nakano, Y.; Ando, H.; Tsujiguchi, H.; Yamashita, T.; Arai, K.; Kaneko, S.; et al. Trajectories of Liver Fibrosis and Gene Expression Profiles in Nonalcoholic Fatty Liver Disease Associated with Diabetes. Diabetes 2023, 72, 1297–1306. [Google Scholar] [CrossRef]
  156. Khairnar, R.; Islam, M.A.; Fleishman, J.; Kumar, S. Shedding light on non-alcoholic fatty liver disease: Pathogenesis, molecular mechanisms, models, and emerging therapeutics. Life Sci. 2023, 312, 121185. [Google Scholar] [CrossRef]
  157. Gastaldelli, A.; Cusi, K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep. 2019, 1, 312–328. [Google Scholar] [CrossRef]
  158. Masuda, K.; Noguchi, S.; Ono, M.; Ochi, T.; Munekage, K.; Okamoto, N.; Suganuma, N.; Saibara, T. High fasting insulin concentrations may be a pivotal predictor for the severity of hepatic fibrosis beyond the glycemic status in non-alcoholic fatty liver disease patients before development of diabetes mellitus. Hepatol. Res. 2017, 47, 983–990. [Google Scholar] [CrossRef] [PubMed]
  159. Noureddin, N.; Noureddin, M.; Singh, A.; Alkhouri, N. Progression of Nonalcoholic Fatty Liver Disease-Associated Fibrosis in a Large Cohort of Patients with Type 2 Diabetes. Dig. Dis. Sci. 2022, 67, 1379–1388. [Google Scholar] [CrossRef] [PubMed]
  160. Paradis, V.; Perlemuter, G.; Bonvoust, F.; Dargere, D.; Parfait, B.; Vidaud, M.; Conti, M.; Huet, S.; Ba, N.; Buffet, C.; et al. High glucose and hyperinsulinemia stimulate connective tissue growth factor expression: A potential mechanism involved in progression to fibrosis in nonalcoholic steatohepatitis. Hepatology 2001, 34 Pt 1, 738–744. [Google Scholar] [CrossRef]
  161. Svegliati-Baroni, G.; Ridolfi, F.; Di Sario, A.; Casini, A.; Marucci, L.; Gaggiotti, G.; Orlandoni, P.; Macarri, G.; Perego, L.; Benedetti, A.; et al. Insulin and insulin-like growth factor-1 stimulate proliferation and type I collagen accumulation by human hepatic stellate cells: Differential effects on signal transduction pathways. Hepatology 1999, 29, 1743–1751. [Google Scholar] [CrossRef]
  162. Carbone, L.J.; Angus, P.W.; Yeomans, N.D. Incretin-based therapies for the treatment of non-alcoholic fatty liver disease: A systematic review and meta-analysis. J. Gastroenterol. Hepatol. 2016, 31, 23–31. [Google Scholar] [CrossRef]
  163. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; LEAN trial team; Abouda, G.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016, 387, 679–690. [Google Scholar] [CrossRef]
  164. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.S.; Harrison, S.A.; NN9931-4296 Investigators. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef]
  165. Musso, G.; Cassader, M.; Paschetta, E.; Gambino, R. Thiazolidinediones and advanced liver fibrosis in nonalcoholic Steatohepatitis: A metaanalysis. JAMA Intern. Med. 2017, 177, 633–640. [Google Scholar] [CrossRef]
  166. Shimizu, M.; Suzuki, K.; Kato, K.; Jojima, T.; Iijima, T.; Murohisa, T.; Iijima, M.; Takekawa, H.; Usui, I.; Hiraishi, H.; et al. Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes Obes. Metab. 2019, 21, 285–292. [Google Scholar] [CrossRef]
  167. Arai, T.; Atsukawa, M.; Tsubota, A.; Mikami, S.; Haruki, U.; Yoshikata, K.; Ono, H.; Kawano, T.; Yoshida, Y.; Tanabe, T.; et al. Antifibrotic effect and long-term outcome of SGLT2 inhibitors in patients with NAFLD complicated by diabetes mellitus. Hepatol. Commun. 2022, 6, 3073–3082. [Google Scholar] [CrossRef]
  168. Thong, V.D.; Quynh, B.T.H. Correlation of Serum Transaminase Levels with Liver Fibrosis Assessed by Transient Elastography in Vietnamese Patients with Nonalcoholic Fatty Liver Disease. Int. J. Gen. Med. 2021, 14, 1349–1355. [Google Scholar] [CrossRef]
  169. Pais, R.; Rusu, E.; Ratziu, V. The impact of obesity and metabolic syndrome on chronic hepatitis B and drug-induced liver disease. Clin. Liver Dis. 2014, 18, 165–178. [Google Scholar] [CrossRef]
  170. Bessone, F.; Dirchwolf, M.; Rodil, M.A.; Razori, M.V.; Roma, M.G. Review article: Drug-induced liver injury in the context of nonalcoholic fatty liver disease—A physiopathological and clinical integrated view. Aliment. Pharmacol. Ther. 2018, 48, 892–913. [Google Scholar] [CrossRef]
  171. Allard, J.; Le Guillou, D.; Begriche, K.; Fromenty, B. Drug-induced liver injury in obesity and nonalcoholic fatty liver disease. Adv. Pharmacol. 2019, 85, 75–107. [Google Scholar]
  172. Massart, J.; Begriche, K.; Moreau, C.; Fromenty, B. Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity. J. Clin. Transl. Res. 2017, 3 (Suppl. S1), 212–232. [Google Scholar]
  173. Cernea, S.; Cahn, A.; Raz, I. Pharmacological management of nonalcoholic fatty liver disease in type 2 diabetes. Expert. Rev. Clin. Pharmacol. 2017, 10, 535–547. [Google Scholar] [CrossRef]
  174. Petroni, M.L.; Brodosi, L.; Bugianesi, E.; Marchesini, G. Management of non-alcoholic fatty liver disease. BMJ 2021, 372, m4747. [Google Scholar] [CrossRef]
  175. Prikhodko, V.A.; Bezborodkina, N.N.; Okovityi, S.V. Pharmacotherapy for Non-Alcoholic Fatty Liver Disease: Emerging Targets and Drug Candidates. Biomedicines 2022, 10, 274. [Google Scholar] [CrossRef] [PubMed]
  176. Zhang, C.Y.; Liu, S.; Yang, M. Treatment of liver fibrosis: Past, current, and future. World J. Hepatol. 2023, 15, 755–774. [Google Scholar] [CrossRef]
  177. Cusi, K.; Orsak, B.; Bril, F.; Lomonaco, R.; Hecht, J.; Ortiz-Lopez, C.; Tio, F.; Hardies, J.; Darland, C.; Musi, N.; et al. Long-Term Pioglitazone Treatment for Patients with Nonalcoholic Steatohepatitis and Prediabetes or Type 2 Diabetes Mellitus: A Randomized Trial. Ann. Intern. Med. 2016, 165, 305–315. [Google Scholar] [CrossRef]
  178. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L.; et al. A Randomized, Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. N. Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef]
  179. A Study of Tirzepatide (LY3298176) in Participants with Nonalcoholic Steatohepatitis (NASH) (SYNERGY-NASH). Available online: https://clinicaltrials.gov/study/NCT04166773?term=NCT04166773&rank=1 (accessed on 10 February 2024).
  180. Dapagliflozin Efficacy and Action in NASH (DEAN). Available online: https://clinicaltrials.gov/study/NCT03723252?term=NCT03723252&rank=1 (accessed on 10 February 2024).
  181. A Phase 3 Study to Evaluate the Efficacy and Safety of MGL-3196 (Resmetirom) in Patients with NASH and Fibrosis (MAESTRO-NASH). Available online: https://clinicaltrials.gov/study/NCT03900429?term=NCT03900429&rank=1 (accessed on 10 February 2024).
  182. A Clinical Study to Evaluate the Efficacy and Safety of Aramchol in Subjects with NASH (ARMOR) (ARMOR). Available online: https://clinicaltrials.gov/study/NCT04104321?term=NCT04104321&rank=1 (accessed on 10 February 2024).
  183. Ratziu, V.; Yilmaz, Y.; Lazas, D.; Friedman, S.L.; Hayardeny, L.; Kadosh, S.; Gorfine, T.; Sanya, A.J. New Data on Aramchol™: Efficacy of a Higher Daily Exposure to Aramchol on Fibrosis Improvement in the ARMOR Study Open-Label Part. EMJ Hepatol. 2022, 10 (Suppl. S1), 2–8. [Google Scholar]
  184. Study to Evaluate the Efficacy, Safety, and Pharmacokinetics of BFKB8488A Compared with Placebo in Participants with Non-Alcoholic Steatohepatitis (BANFF). Available online: https://clinicaltrials.gov/study/NCT04171765?term=NCT04171765&rank=1 (accessed on 10 February 2024).
  185. Harrison, S.A.; Ruane, P.J.; Freilich, B.L.; Neff, G.; Patil, R.; Behling, C.A.; Hu, C.; Fong, E.; de Temple, B.; Tillman, E.J.; et al. Efruxifermin in non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled, phase 2a trial. Nat. Med. 2021, 27, 1262–1271. [Google Scholar] [CrossRef]
  186. Dasatinib and Quercetin to Treat Fibrotic Non-alcoholic Fatty Liver Disease. Available online: https://clinicaltrials.gov/study/NCT05506488?term=NCT05506488&rank=1 (accessed on 11 February 2024).
  187. Schwabe, R.F.; Tabas, I.; Pajvani, U.B. Mechanisms of Fibrosis Development in Nonalcoholic Steatohepatitis. Gastroenterology 2020, 158, 1913–1928. [Google Scholar] [CrossRef]
  188. Tacke, F.; Puengel, T.; Loomba, R.; Friedman, S.L. An integrated view of anti-inflammatory and antifibrotic targets for the treatment of NASH. J. Hepatol. 2023, 79, 552–566. [Google Scholar] [CrossRef]
  189. Tacke, F.; Weiskirchen, R. Non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH)-related liver fibrosis: Mechanisms, treatment and prevention. Ann. Transl. Med. 2021, 9, 729. [Google Scholar] [CrossRef] [PubMed]
  190. Gilgenkrantz, H.; Mallat, A.; Moreau, R.; Lotersztajn, S. Targeting cell-intrinsic metabolism for antifibrotic therapy. J. Hepatol. 2021, 74, 1442–1454. [Google Scholar] [CrossRef] [PubMed]
  191. Mohammadzadeh, G.; Afarin, R.; Bavarsad, S.S.; Aslani, F.; Zadeh, S.A.; Shakerian, E. Comparison of the effects of cholesterol, palmitic acid, and glucose on activation of human hepatic stellate cells to induce liver fibrosis. J. Diabetes Metab. Disord. 2022, 21, 1531–1538. [Google Scholar] [CrossRef] [PubMed]
  192. Sugimoto, R.; Enjoji, M.; Kohjima, M.; Tsuruta, S.; Fukushima, M.; Iwao, M.; Sonta, T.; Kotoh, K.; Inoguchi, T.; Nakamuta, M. High glucose stimulates hepatic stellate cells to proliferate and to produce collagen through free radical production and activation of mitogen-activated protein kinase. Liver Int. 2005, 25, 1018–1026. [Google Scholar] [CrossRef] [PubMed]
  193. Wu, L.K.; Liu, Y.C.; Ma, G.; Shi, L.L.; He, X.M. High levels of glucose promote the activation of hepatic stellate cells via the p38-mitogen-activated protein kinase signal pathway. Genet. Mol. Res. 2016, 15, gmr.15038419. [Google Scholar] [CrossRef] [PubMed]
  194. Trivedi, P.; Wang, S.; Friedman, S.L. The Power of Plasticity-Metabolic Regulation of Hepatic Stellate Cells. Cell Metab. 2021, 33, 242–257. [Google Scholar] [CrossRef]
  195. Hernández-Gea, V.; Friedman, S.L. Autophagy fuels tissue fibrogenesis. Autophagy 2012, 8, 849–850. [Google Scholar] [CrossRef]
  196. Ioannou, G.N.; Subramanian, S.; Chait, A.; Haigh, W.G.; Yeh, M.M.; Farrell, G.C.; Lee, S.P.; Savard, C. Cholesterol crystallization within hepatocyte lipid droplets and its role in murine NASH. J. Lipid Res. 2017, 58, 1067–1079. [Google Scholar] [CrossRef]
  197. Leroux, A.; Ferrere, G.; Godie, V.; Cailleux, F.; Renoud, M.L.; Gaudin, F.; Naveau, S.; Prévot, S.; Makhzami, S.; Perlemuter, G.; et al. Toxic lipids stored by Kupffer cells correlates with their pro-inflammatory phenotype at an early stage of steatohepatitis. J. Hepatol. 2012, 57, 141–149. [Google Scholar] [CrossRef]
  198. Kazankov, K.; Jørgensen, S.M.D.; Thomsen, K.L.; Møller, H.J.; Vilstrup, H.; George, J.; Schuppan, D.; Grønbæk, H. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 145–159. [Google Scholar] [CrossRef]
Figure 1. Factors associated with the progression/regression of NAFLD-associated liver fibrosis (see text for details).
Figure 1. Factors associated with the progression/regression of NAFLD-associated liver fibrosis (see text for details).
Life 14 00272 g001
Table 1. Summary of main drugs with potential benefits in terms of NAFLD-related fibrosis (PPAR: peroxisome proliferator activated receptor; RCT: randomized controlled trial; pbo: placebo; NAS: non-alcoholic fatty liver disease activity score; TD: treatment difference; SAF-A: the activity part of the steatosis, activity, fibrosis [SAF] scoring system that incorporates scores for ballooning and inflammation; GLP-1 RA: glucagon-like peptide-1 receptor agonist; GIP: glucose-dependent insulinotropic polypeptide; RR: relative risk; SGLT 2: sodium–glucose cotransporter 2; N/A: not available; wk: week; THR: thyroid hormone receptor; SCD1: stearoyl-CoA desaturase; BID: twice a day; FGF: fibroblast growth factor; HFF: hepatic fat fraction; ELF: enhanced liver fibrosis; [] is presented as 95% confidence interval) [158,177,178,179,180,181,182,183,184,185,186].
Table 1. Summary of main drugs with potential benefits in terms of NAFLD-related fibrosis (PPAR: peroxisome proliferator activated receptor; RCT: randomized controlled trial; pbo: placebo; NAS: non-alcoholic fatty liver disease activity score; TD: treatment difference; SAF-A: the activity part of the steatosis, activity, fibrosis [SAF] scoring system that incorporates scores for ballooning and inflammation; GLP-1 RA: glucagon-like peptide-1 receptor agonist; GIP: glucose-dependent insulinotropic polypeptide; RR: relative risk; SGLT 2: sodium–glucose cotransporter 2; N/A: not available; wk: week; THR: thyroid hormone receptor; SCD1: stearoyl-CoA desaturase; BID: twice a day; FGF: fibroblast growth factor; HFF: hepatic fat fraction; ELF: enhanced liver fibrosis; [] is presented as 95% confidence interval) [158,177,178,179,180,181,182,183,184,185,186].
Drug Name/
Mechanism of
Action
Study/Clinical Trial Ref.Study Population/NumberPrimary ObjectiveMain Results (If Available)
Monotherapy
Pioglitazone/PPAR-γ agonist; insulin sensitizerRCT (pioglitazone
45 mg/day vs. pbo), NCT00994682 [177]
T2DM/prediabetes and biopsy-proven NASH
(n = 101)
Reduction of ≥2 NAS points in two histologic categories without worsening of fibrosis.Primary outcome:
TD: 41 [23; 59] percentage points;
p < 0.001.
Additional outcome:
mean change in fibrosis score:
TD: −0.5 [−0.9; 0.0]; p = 0.039.
Lanifibranor/pan-PPAR agonistRCT (lanifibranor 1200 mg/800 mg per day or pbo), NCT03008070 [178]Non-cirrhotic, highly active NASH (n = 247)Decrease of ≥2 points in SAF-A score without worsening of fibrosis.Primary outcome:
RR: 1.7 [1.2; 2.3]; p = 0.007 (1200 mg dose vs. pbo);
RR: 1.5 [1.0; 2.1]; p = 0.07 (800 mg dose vs. pbo).
Additional outcome:
Improvement in fibrosis stage of ≥1 without worsening of NASH:
RR: 1.7 [1.2; 2.5] (1200 mg dose vs. pbo);
RR: 1.2 [0.7; 1.9] (800 mg dose vs. pbo).
Liraglutide/
GLP-1 RA
RCT (liraglutide
1.8 mg/day vs. pbo), NCT01237119 [158]
Biopsy-proven NASH
(n = 52; 17 with T2DM)
Resolution of definite NASH with no worsening of fibrosis.Primary outcome:
RR: 4.3 [1.0; 17.7]; p = 0.019.
Additional results:
patients with worsening of fibrosis:
mean change from baseline vs. pbo: 0.2 [0.1; 1.0]; p = 0.04.
Tirzepatide/
Dual GLP-1 and GIP RA
RCT (tirzepatide 5 mg/10 mg/15 mg per wk vs. pbo), NCT04166773 [179]Biopsy-proven NASH, and stage F2/3 fibrosis, or without T2DM
(n = 196 estimated)
Percentage of participants with absence of NASH with no worsening of fibrosis.
Secondary outcomes:
percentage of participants with ≥1 point decrease in fibrosis stage with no worsening of NASH;
percentage of participants with ≥1 point increase in fibrosis stage with no worsening of NASH.
N/A; ongoing; phase 2
Dapagliflozin/
SGLT 2 inhibitors
RCT (dapagliflozin
10 mg/day vs. pbo), NCT03723252 [180]
Biopsy-proven NASH
(n = 148 estimated)
Improvement in scored liver histology over 12 months.
Secondary outcome:
change in fibrosis score.
N/A; ongoing; phase 3
Resmetirom/
THR β-selective agonist
RCT (resmetirom (MGL-3196)
80 mg/100 mg per day vs. pbo), NCT03900429 [181]
NASH fibrosis
(n = 1759 enrolled)
Proportion with resolution of NASH associated with ≥2-point reduction in NAS without worsening of fibrosis stage OR
proportion with ≥1-point improvement in fibrosis stage with no worsening of NAS.
N/A; ongoing; phase 3
Aramchol/
partial inhibitor of hepatic SCD1
RCT (aramchol
300 mg BID or pbo), double-blind and open-label; NCT04104321 [182,183]
Biopsy-proven NASH with fibrosis stage F2/3, overweight/obesity, and prediabetes, or T2DM (for double-blind) (150 for open-label; n = 2000 estimated for double-blind)Proportion of subjects with improvement in liver fibrosis ≥1 and no worsening of NASH.
Proportion of subjects with resolution of NASH and no worsening of liver fibrosis.
Open-label interim analysis:
Primary outcome:
60.0% had fibrosis improvement of ≥1 stage (of first 20 patients).
N/A; phase 3
BFKB8488A/
FGF receptor 1/Klothoβ agonist
RCT (individualized or fixed doses of BFKB8488A vs. pbo), NCT04171765 [184]Biopsy-proven NASH with stage F2/3 fibrosis, and liver fat ≥8%Proportion of participants with NASH resolution without worsening of fibrosis.
Secondary outcome:
proportion of participants with improvement in liver fibrosis of ≥1 stage and no worsening of NASH.
N/A; phase 2
Efruxifermin/
Fc-FGF21 fusion protein; FGF receptor agonist
RCT (efruxifermin 28 mg/50 mg/70 mg per wk vs. pbo), NCT03976401 [185]Biopsy-proven NASH and ≥10% liver fat content
(n = 80)
Absolute change from baseline in HFF measured by magnetic resonance imaging.
Secondary outcomes:
change from baseline in liver stiffness;
change from baseline in non-invasive biomarkers including liver fibrosis.
Primary outcome:
absolute changes in HFF: −12.3%, −13.4%, and −14.1% (28, 50, and
70 mg) vs. 0.3% (pbo); p < 0.0001.
Additional outcomes:
reduction in ELF scores (p = 0.0008 (28 mg), p = 0.0005 (50 mg), and p = 0.03 (70 mg) vs. pbo);
55% (across all efruxifermin arms with liver biopsies) had a fibrosis improvement of ≥1 stage (no statistical analysis vs. pbo).
Combination therapy
Dasatinib and Quercetin/tyrosine kinase inhibitor and flavonoidRCT (dasatinib (100 mg/day) plus quercetin (1000 mg/day) on three consecutive days for three consecutive wks), NCT05506488 [186]Biopsy-proven NAFLD with stage >F2 fibrosis, but no cirrhosis (n = 30 estimated)Improvement of fibrosis with at least 1-point without worsening of fibrosis and NAFLD score based on histology.N/A; ongoing; phase 1, 2
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cernea, S. NAFLD Fibrosis Progression and Type 2 Diabetes: The Hepatic–Metabolic Interplay. Life 2024, 14, 272. https://doi.org/10.3390/life14020272

AMA Style

Cernea S. NAFLD Fibrosis Progression and Type 2 Diabetes: The Hepatic–Metabolic Interplay. Life. 2024; 14(2):272. https://doi.org/10.3390/life14020272

Chicago/Turabian Style

Cernea, Simona. 2024. "NAFLD Fibrosis Progression and Type 2 Diabetes: The Hepatic–Metabolic Interplay" Life 14, no. 2: 272. https://doi.org/10.3390/life14020272

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop