Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (212)

Search Parameters:
Keywords = metabolic-dysfunction-associated liver steatohepatitis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 2024 KiB  
Article
Oxy210 Inhibits Hepatic Expression of Senescence-Associated, Pro-Fibrotic, and Pro-Inflammatory Genes in Mice During Development of MASH and in Hepatocytes In Vitro
by Feng Wang, Simon T. Hui, Frank Stappenbeck, Dorota Kaminska, Aldons J. Lusis and Farhad Parhami
Cells 2025, 14(15), 1191; https://doi.org/10.3390/cells14151191 - 2 Aug 2025
Viewed by 252
Abstract
Background: Senescence, a state of permanent cell cycle arrest, is a complex cellular phenomenon closely affiliated with age-related diseases and pathological fibrosis. Cellular senescence is now recognized as a significant contributor to organ fibrosis, largely driven by transforming growth factor beta (TGF-β) signaling, [...] Read more.
Background: Senescence, a state of permanent cell cycle arrest, is a complex cellular phenomenon closely affiliated with age-related diseases and pathological fibrosis. Cellular senescence is now recognized as a significant contributor to organ fibrosis, largely driven by transforming growth factor beta (TGF-β) signaling, such as in metabolic dysfunction-associated steatohepatitis (MASH), idiopathic pulmonary fibrosis (IPF), chronic kidney disease (CKD), and myocardial fibrosis, which can lead to heart failure, cystic fibrosis, and fibrosis in pancreatic tumors, to name a few. MASH is a progressive inflammatory and fibrotic liver condition that has reached pandemic proportions, now considered the largest non-viral contributor to the need for liver transplantation. Methods: We previously studied Oxy210, an anti-fibrotic and anti-inflammatory, orally bioavailable, oxysterol-based drug candidate for MASH, using APOE*3-Leiden.CETP mice, a humanized hyperlipidemic mouse model that closely recapitulates the hallmarks of human MASH. In this model, treatment of mice with Oxy210 for 16 weeks caused significant amelioration of the disease, evidenced by reduced hepatic inflammation, lipid deposition, and fibrosis, atherosclerosis and adipose tissue inflammation. Results: Here we demonstrate increased hepatic expression of senescence-associated genes and senescence-associated secretory phenotype (SASP), correlated with the expression of pro-fibrotic and pro-inflammatorygenes in these mice during the development of MASH that are significantly inhibited by Oxy210. Using the HepG2 human hepatocyte cell line, we demonstrate the induced expression of senescent-associated genes and SASP by TGF-β and inhibition by Oxy210. Conclusions: These findings further support the potential therapeutic effects of Oxy210 mediated in part through inhibition of senescence-driven hepatic fibrosis and inflammation in MASH and perhaps in other senescence-associated fibrotic diseases. Full article
Show Figures

Graphical abstract

34 pages, 11716 KiB  
Article
UPLC-MS/MS Metabolomics Reveals Babao Dan’s Mechanisms in MASH Treatment with Integrating Network Pharmacology and Molecular Docking
by Shijiao Zhang, Yanding Su, Ao Han, He Qi, Jiade Zhao and Xiangjun Qiu
Pharmaceuticals 2025, 18(8), 1111; https://doi.org/10.3390/ph18081111 - 25 Jul 2025
Viewed by 237
Abstract
Background: Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive disease that easily develops into cirrhosis and hepatocellular carcinoma, but its pathogenesis is not clear, and most therapeutic drugs have obvious limitations. However, Babao Dan (BBD) has a good therapeutic effect on liver disease, [...] Read more.
Background: Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive disease that easily develops into cirrhosis and hepatocellular carcinoma, but its pathogenesis is not clear, and most therapeutic drugs have obvious limitations. However, Babao Dan (BBD) has a good therapeutic effect on liver disease, but its treatment mechanism is still to be studied. Therefore, we further investigated the mechanism of BBD in treating MASH. Methods: We predicted BBD-related targets through network pharmacology and further verified the binding ability of BBD-related targets through molecular docking. We also detected relevant indicators before and after model treatment, as well as metabolomics analysis and identification of the mechanism of action of BBD on MASH. Results: Through network pharmacology methods, 158 key cross targets and the top 10 core targets were identified, and it was determined that the PI3K-AKT signaling pathway plays an important regulatory role in the treatment of MASH with BBD. The molecular docking results indicate that the representative compounds quercetin and 17 Beta Estradiol have good binding activity with five core targets. Metabolomics has identified four metabolic biomarkers, such as Piceid, and it is determined that the key pathway for BBD treatment of MASH is the bile secretion pathway. Conclusions: BBD effectively treats MASH by modulating Piceid and other biomarkers, targeting ESR1 and other core proteins via quercetin and 17-beta-estradiol, and regulating the PI3K-AKT and bile secretion pathways to alleviate liver injury. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

17 pages, 4120 KiB  
Article
Albumin Reduces Hepatic Steatosis and Inflammation in High-Fat-Diet-Fed Mice
by Claire Rennie, Sheila Donnelly and Kristine McGrath
Int. J. Mol. Sci. 2025, 26(15), 7156; https://doi.org/10.3390/ijms26157156 - 24 Jul 2025
Viewed by 217
Abstract
There are currently no approved therapeutic treatments targeting metabolic dysfunction-associated steatotic liver disease (MASLD). Albumin, a liver-produced plasma protein with anti-inflammatory and antioxidant properties, is reduced in advanced liver disease. Considering the role of chronic obesity-induced inflammation in MASLD pathogenesis, we investigated whether [...] Read more.
There are currently no approved therapeutic treatments targeting metabolic dysfunction-associated steatotic liver disease (MASLD). Albumin, a liver-produced plasma protein with anti-inflammatory and antioxidant properties, is reduced in advanced liver disease. Considering the role of chronic obesity-induced inflammation in MASLD pathogenesis, we investigated whether albumin administration could prevent disease progression to metabolic dysfunction-associated steatohepatitis (MASH). MASLD was induced in mice using a high-fat and high-cholesterol (PC) treatment for 8 weeks, followed by treatment with bovine serum albumin (BSA; 0.8 mg/kg) every three days for another 8 weeks. This regimen prevented time-dependent weight gain, regardless of diet, with 57% and 27% reductions in mice fed a standard chow (Std Chow) or PC diet, respectively. Further, supplementation reduced nuclear factor kappa B (NF-κB) activation by 2.8-fold (p = 0.0328) in PC-fed mice, consistent with albumin’s known anti-inflammatory properties. Unexpectedly, albumin also reduced hepatic neutral lipid accumulation and circulating non-esterified fatty acids. While PC-fed mice did not exhibit full progression to MASH, albumin treatment significantly increased hepatic matrix metalloproteinase-2 expression, suggesting the inhibition of early fibrotic signalling. While further studies are needed to elucidate the underlying mechanisms, these findings offer new insight into the potential of albumin, either alone or in combination with other therapies, to reduce hepatic steatosis in MASLD. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Graphical abstract

30 pages, 2885 KiB  
Review
Targeting Lipophagy in Liver Diseases: Impact on Oxidative Stress and Steatohepatitis
by Jin Seok Hwang, Trang Huyen Lai and Deok Ryong Kim
Antioxidants 2025, 14(8), 908; https://doi.org/10.3390/antiox14080908 - 24 Jul 2025
Viewed by 540
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a range of liver conditions, from simple hepatic steatosis to its more severe inflammatory form known as metabolic dysfunction-associated steatohepatitis (MASH). Despite its growing clinical significance and association with cirrhosis and cancer, there are currently few [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a range of liver conditions, from simple hepatic steatosis to its more severe inflammatory form known as metabolic dysfunction-associated steatohepatitis (MASH). Despite its growing clinical significance and association with cirrhosis and cancer, there are currently few pharmacological treatments available for MASLD, highlighting the urgent need for new therapeutic strategies. This narrative review aims to elucidate the molecular mechanisms of lipophagy in MASLD progression, emphasizing how its dysfunction contributes to hepatic steatosis and lipotoxicity. We also explore the intersection of lipophagy failure with oxidative stress and inflammation in the liver, focusing on key signaling pathways, such as mTORC1 and AMPK, and discuss the therapeutic potential of targeting these pathways by systematically reviewing the literature from PubMed, Scopus, and Google Scholar databases. Recent studies suggest that lipophagy, the selective autophagic degradation of lipid droplets, is crucial for maintaining hepatic lipid homeostasis. Indeed, some vital components of the lipophagy machinery seem to be functionally inhibited in MASLD, resulting in the accumulation of intracellular triacylglycerol (TAG), lipotoxicity, and subsequent oxidative stress, all of which contribute to disease progression. In summary, impaired lipophagy is a central pathological mechanism in MASLD, making it an important therapeutic target. A deeper understanding of these mechanisms may offer new strategic insights for combating the progression of MASLD/MASH. Full article
(This article belongs to the Special Issue Oxidative Stress and Liver Disease)
Show Figures

Figure 1

28 pages, 1362 KiB  
Review
Multifaceted Interactions Between Bile Acids, Their Receptors, and MASH: From Molecular Mechanisms to Clinical Therapeutics
by Xuan Tang, Yuanjiao Zhou, Li Xia, Xiulian Lin, Yao Zhu, Menghan Chen, Jiayao Wang and Yamei Li
Molecules 2025, 30(15), 3066; https://doi.org/10.3390/molecules30153066 - 22 Jul 2025
Viewed by 387
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) represents a critical hepatic manifestation within the broader spectrum of metabolic syndrome. The pathogenesis of MASH is characterized by disruptions in lipid metabolism, inflammation, and fibrosis. Bile acids and their receptors are integral to the progression of MASH, primarily [...] Read more.
Metabolic dysfunction-associated steatohepatitis (MASH) represents a critical hepatic manifestation within the broader spectrum of metabolic syndrome. The pathogenesis of MASH is characterized by disruptions in lipid metabolism, inflammation, and fibrosis. Bile acids and their receptors are integral to the progression of MASH, primarily through their regulatory influence on the metabolic networks of the gut–liver axis. This review offers a comprehensive and systematic examination of the molecular mechanisms underlying bile acid biosynthesis, metabolic dysregulation, and receptor signaling anomalies in MASH. Furthermore, it explores the translational potential of these insights into clinical therapies. Bile acids and their receptors emerge as pivotal therapeutic targets for MASH. Future research should focus on an in-depth analysis of dynamic regulatory mechanisms and the optimization of multi-target combination therapies, thereby paving the way for significant clinical advancements. Full article
(This article belongs to the Special Issue Chemical Biology in Asia—Second Edition)
Show Figures

Figure 1

13 pages, 891 KiB  
Review
The Role of Aldosterone and the Mineralocorticoid Receptor in Metabolic Dysfunction-Associated Steatotic Liver Disease
by Mohammed Barigou, Imran Ramzan and Dionysios V. Chartoumpekis
Biomedicines 2025, 13(8), 1792; https://doi.org/10.3390/biomedicines13081792 - 22 Jul 2025
Viewed by 345
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is one of the fastest-growing hepatic disorders worldwide. During its natural course, MASLD tends to progress from isolated steatosis of the liver to Metabolic Dysfunction-Associated Alcoholic Steatohepatitis (MASH), advanced fibrosis, and finally cirrhosis, with the risk of [...] Read more.
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is one of the fastest-growing hepatic disorders worldwide. During its natural course, MASLD tends to progress from isolated steatosis of the liver to Metabolic Dysfunction-Associated Alcoholic Steatohepatitis (MASH), advanced fibrosis, and finally cirrhosis, with the risk of developing hepatocellular carcinoma (HCC). Although frequently related to overweight or obesity and other components of the metabolic syndrome (MS), MASLD can also be present in individuals without such risk factors. The mechanisms leading to MASLD are incompletely elucidated and may involve many proinflammatory and pro-fibrotic pathways, disrupted biliary acid homeostasis, and gut microbiota dysbiosis. Aldosterone and its interaction with the mineralocorticoid receptor (MR) are thought to participate in the pathogenesis of MASLD through the modulation of inflammation and fibrosis. Remarkably, blockade of the MR in experimental models was shown to improve MASH and fibrosis through mechanisms that need further characterization. So far, however, few clinical studies have explored the effect of MR blockade in the management of MASH and associated fibrosis. This review is intended to summarize the recent animal and human data concerning the interaction between MR pathways and MASLD. Full article
(This article belongs to the Special Issue Novel Insights into Liver Metabolism)
Show Figures

Figure 1

33 pages, 1553 KiB  
Review
Multifaceted Human Antigen R (HuR): A Key Player in Liver Metabolism and MASLD
by Natalie Eppler, Elizabeth Jones, Forkan Ahamed and Yuxia Zhang
Livers 2025, 5(3), 33; https://doi.org/10.3390/livers5030033 - 21 Jul 2025
Viewed by 498
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become the leading cause of chronic liver disease worldwide, affecting approximately 25–30% of the global adult population and highlighting the urgent need for effective therapeutics and prevention strategies. MASLD is characterized by excessive hepatic lipid accumulation [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become the leading cause of chronic liver disease worldwide, affecting approximately 25–30% of the global adult population and highlighting the urgent need for effective therapeutics and prevention strategies. MASLD is characterized by excessive hepatic lipid accumulation and can progress, in a subset of patients, to metabolic dysfunction-associated steatohepatitis (MASH), a pro-inflammatory and pro-fibrotic condition associated with increased risk of liver cirrhosis and hepatocellular carcinoma. Although the molecular drivers of MASLD progression remain incompletely understood, several key metabolic pathways—such as triglyceride handling, cholesterol catabolism, bile acid metabolism, mitochondrial function, and autophagy—are consistently dysregulated in MASLD livers. This narrative review summarizes primary literature and highlights insights from recent reviews on the multifaceted role of the mRNA-binding protein Human antigen R (HuR) in the post-transcriptional regulation of critical cellular processes, including nutrient metabolism, cell survival, and stress responses. Emerging evidence underscores HuR’s essential role in maintaining liver homeostasis, particularly under metabolic stress conditions characteristic of MASLD, with hepatocyte-specific HuR depletion associated with exacerbated disease severity. Moreover, comorbid conditions such as obesity, type 2 diabetes mellitus, and cardiovascular disease not only exacerbate MASLD progression but also involve HuR dysregulation in extrahepatic tissues, further contributing to liver dysfunction. A deeper understanding of HuR-regulated post-transcriptional networks across metabolic organs may enable the development of targeted therapies aimed at halting or reversing MASLD progression. Full article
Show Figures

Figure 1

16 pages, 3260 KiB  
Article
Rifaximin Attenuates Liver Fibrosis and Hepatocarcinogenesis in a Rat MASH Model by Suppressing the Gut–Liver Axis and Epiregulin–IL-8-Associated Angiogenesis
by Naoki Nishimura, Kosuke Kaji, Norihisa Nishimura, Junichi Hanatani, Tatsuya Nakatani, Masafumi Oyama, Akihiko Shibamoto, Yuki Tsuji, Koh Kitagawa, Shinya Sato, Tadashi Namisaki, Satoru Tamaoki and Hitoshi Yoshiji
Int. J. Mol. Sci. 2025, 26(14), 6710; https://doi.org/10.3390/ijms26146710 - 12 Jul 2025
Viewed by 400
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disease linked to fibrosis and hepatocellular carcinoma (HCC). Gut-derived lipopolysaccharide (LPS) promotes hepatic inflammation, fibrosis, and angiogenesis through toll-like receptor 4 (TLR4) signaling. This study examined the effects of rifaximin, a non-absorbable, gut-targeted antibiotic, on [...] Read more.
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disease linked to fibrosis and hepatocellular carcinoma (HCC). Gut-derived lipopolysaccharide (LPS) promotes hepatic inflammation, fibrosis, and angiogenesis through toll-like receptor 4 (TLR4) signaling. This study examined the effects of rifaximin, a non-absorbable, gut-targeted antibiotic, on MASH-related liver fibrosis and early hepatocarcinogenesis, with a focus on the LPS–epiregulin–IL-8–angiogenesis axis.MASH was induced in Fischer 344 rats using a choline-deficient, L-amino acid-defined high-fat diet (CDAHFD). Rifaximin (30 mg/kg/day) was orally administered for 12 weeks. Liver histology, gene expression, intestinal permeability, LPS levels, and angiogenic markers were evaluated. Rifaximin reduced hepatic inflammation, fibrosis, hydroxyproline content, and fibrogenic gene expression. The number and size of GST-P-positive preneoplastic lesions and proliferation-related genes were decreased. Portal LPS levels and Kupffer cell activation declined, with downregulation of Lbp, Cd14, Tlr4, and inflammatory cytokines. Rifaximin decreased hepatic epiregulin and IL-8 expression, attenuated CD34-positive neovascularization, and suppressed proangiogenic gene expression, accompanied by improved intestinal barrier function and reduced gut permeability. Rifaximin mitigates MASH progression by restoring gut barrier integrity, limiting LPS translocation, and inhibiting fibrogenic and angiogenic pathways. These results suggest its potential as a chemopreventive agent in MASH-related hepatocarcinogenesis. Full article
(This article belongs to the Special Issue Liver Diseases: From Molecular Basis to Potential Therapy)
Show Figures

Figure 1

14 pages, 1915 KiB  
Article
Effects of Prebiotics Inulin and Oat β-Glucan on Colonic Architecture and Hepatic Proteome in Mice with Circadian-Disruption-Aggravated Metabolic Dysfunction-Associated Steatohepatitis
by Nelson Kei, Kam Kuen Cheung, Ka Lee Ma, Tsz Kwan Yau, Susana Lauw, Xing Kang, Kiwi Wai Yan Sun, Yu Wang, Vincent Wai Sun Wong, Sunny Hei Wong and Peter Chi Keung Cheung
Nutrients 2025, 17(13), 2245; https://doi.org/10.3390/nu17132245 - 7 Jul 2025
Viewed by 522
Abstract
Background: Circadian disruption (CD) aggravates metabolic dysfunction-associated steatohepatitis (MASH), but supplementation with prebiotics inulin and oat β-glucan may mitigate its effects. However, their impact on colonic architecture and hepatic proteome remains unclear. Objectives: We aimed to investigate the effects of prebiotics inulin and [...] Read more.
Background: Circadian disruption (CD) aggravates metabolic dysfunction-associated steatohepatitis (MASH), but supplementation with prebiotics inulin and oat β-glucan may mitigate its effects. However, their impact on colonic architecture and hepatic proteome remains unclear. Objectives: We aimed to investigate the effects of prebiotics inulin and oat β-glucan on colonic architecture and hepatic proteome in mice with CD-aggravated MASH. Methods: CD was induced by weekly reversal of the light–dark cycle to simulate shift work. Male C57BL/6J mice were subjected to non-shifted chow, non-shifted fructose, palmitate, cholesterol, and trans-fat (FPC) diet, shifted chow, or shifted FPC diet (SFPC) for 26 weeks. Prebiotics inulin and oat β-glucan supplementation were provided to the SFPC group in the final 10 weeks. Distal colon and serum samples were collected for histological examination and endotoxemia evaluation, respectively. Liver samples were collected for proteomic mass spectrometry analysis. Results: Mice with CD-aggravated MASH were found with colonic crypt loss and a distinct hepatic proteome structure compared to mice with non-CD MASH. Notably, inulin showed better effects than oat β-glucan in preserving colonic crypts in mice with CD-aggravated MASH. Furthermore, inulin supplementation restored the hepatic proteome structure similar to that of non-CD MASH mice, a benefit not observed with oat β-glucan. Conclusions: Given our prior findings showing oat β-glucan’s superior ability to enrich gut bacterial species associated with MASH improvement under CD, this study highlights inulin’s unique benefits for colonic architecture and hepatic proteome regulation in CD-aggravated MASH. Full article
Show Figures

Figure 1

13 pages, 3483 KiB  
Article
Levels of Growth Differentiation Factor 15 Correlated with Metabolic Dysfunction-Associated Steatotic Liver Disease in Children
by Antonella Mosca, Maria Rita Braghini, Giulia Andolina, Cristiano De Stefanis, Lucia Cesarini, Anna Pastore, Donatella Comparcola, Lidia Monti, Paola Francalanci, Clara Balsano, Andrea Pietrobattista, Anna Alisi and Nadia Panera
Int. J. Mol. Sci. 2025, 26(13), 6486; https://doi.org/10.3390/ijms26136486 - 5 Jul 2025
Viewed by 429
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic progressive hepatopathy in children, and the identification of non-invasive biomarkers is urgently needed. Growth differentiation factor 15 (GDF15) was associated with MASLD in adults. In this study, we investigated the circulating and [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic progressive hepatopathy in children, and the identification of non-invasive biomarkers is urgently needed. Growth differentiation factor 15 (GDF15) was associated with MASLD in adults. In this study, we investigated the circulating and hepatic levels of GDF15 and their association with liver damage in pediatric MASLD and in a murine model. This observational study included 158 children with biopsy-proven MASLD. Patients with MASLD were categorized into two groups based on steatohepatitis (MASH) presence and evaluated for GDF15 circulating levels, while GDF15 hepatic levels were assessed only in a subset of patients. Children with MASLD exhibited higher levels of circulating GDF15 compared to the controls. Moreover, the MASH subgroup had significantly higher values of GDF15 compared to the Not-MASH subgroup. The GDF15 levels in the MASH subgroup showed a positive correlation with fibrosis. Finally, the hepatic expression of the GDF15 gene correlated with GDF15 circulating levels and with the hepatic expression of the COL1A1 and COL3A1 genes in 15 children with MASLD. In conclusion, our study demonstrated that GDF15 levels are associated with liver damage, reinforcing the potential role of GDF15 as a biomarker for MASLD-related fibrosis in children. Full article
(This article belongs to the Special Issue Steatotic Liver Disease: From Bench to Bedside and Back)
Show Figures

Graphical abstract

32 pages, 1794 KiB  
Review
Food Nutrients and Bioactive Compounds for Managing Metabolic Dysfunction-Associated Steatotic Liver Disease: A Comprehensive Review
by Erdenetsogt Dungubat, Kohei Fujikura, Masahiko Kuroda, Toshio Fukusato and Yoshihisa Takahashi
Nutrients 2025, 17(13), 2211; https://doi.org/10.3390/nu17132211 - 3 Jul 2025
Viewed by 975
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH), are growing global health concerns. However, pharmacological therapies for MASLD/MASH have not yet been established. Dietary interventions and their bioactive components have been explored as strategies to mitigate MASLD [...] Read more.
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH), are growing global health concerns. However, pharmacological therapies for MASLD/MASH have not yet been established. Dietary interventions and their bioactive components have been explored as strategies to mitigate MASLD and MASH progression. Although specific nutrients and bioactive compounds have exhibited potential therapeutic benefits, they also exacerbate adverse outcomes. In this comprehensive review, we synthesize the protective and exacerbating or sometimes dual effects of key macronutrients, including fatty acids (saturated, unsaturated, and trans fats) and carbohydrates (fructose, glucose, and sucrose), and bioactive compounds and micronutrients, in the context of MASLD management. The evidence suggests that coffee-derived compounds, such as caffeine and chlorogenic acid, may attenuate liver injury. However, the effects on MASLD severity are inconsistent. Diets high in saturated fatty acids exacerbate MASLD pathogenesis, whereas moderate intake (7–10% of total energy) may confer metabolic benefits. Other bioactive compounds and micronutrients have been explored for their diverse roles in hepatic lipid metabolism and insulin sensitivity. Although current evidence supports the therapeutic potential of specific dietary nutrients and bioactive compounds in the management of MASLD, inconsistencies in results highlight the need for more robust, well-controlled studies, including clinical trials, to clarify the preventive and therapeutic standards for balanced food interventions in MASLD management. In particular, well-designed clinical trials are necessary before clinical application. Although this is a narrative review and the literature retrieval may be biased, we covered a wide variety of substances. Full article
Show Figures

Figure 1

13 pages, 532 KiB  
Systematic Review
The Comparative Safety and Efficacy of Resmetirom and Semaglutide in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A Systematic Review
by Jahnavi Udaikumar, Rithish Nimmagadda, Vindhya Vasini Lella, Kesava Manikanta Achuta, Satwik Kuppili, Suraj Reddy Avula and Raiya Sarwar
Pharmacoepidemiology 2025, 4(3), 14; https://doi.org/10.3390/pharma4030014 - 27 Jun 2025
Viewed by 751
Abstract
Introduction: Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly encompassed under nonalcoholic fatty liver disease (NAFLD), is a growing global health burden associated with progression to cirrhosis and hepatocellular carcinoma. Resmetirom, a thyroid hormone receptor-β (THR-β) agonist, and semaglutide, a glucagon-like peptide-1 receptor [...] Read more.
Introduction: Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly encompassed under nonalcoholic fatty liver disease (NAFLD), is a growing global health burden associated with progression to cirrhosis and hepatocellular carcinoma. Resmetirom, a thyroid hormone receptor-β (THR-β) agonist, and semaglutide, a glucagon-like peptide-1 receptor agonist (GLP-1 RA), have emerged as promising agents targeting distinct metabolic and inflammatory pathways. This systematic review compares the safety and efficacy of resmetirom and semaglutide in MASLD. Methods: We conducted a comprehensive search of PubMed, Embase, and Google Scholar for randomized controlled trials and clinical studies published between January 2014 and April 2025, following PRISMA guidelines. Studies assessing the efficacy and safety of resmetirom and/or semaglutide in MASLD or NASH were included. Data extraction was performed by two independent reviewers, and a narrative synthesis was undertaken due to the heterogeneity in study design and outcome measures. Results: Fourteen studies encompassing over 4500 patients were analyzed. Resmetirom demonstrated consistent reductions in hepatic fat (≥30% in >50% of patients) and improvements in fibrosis (≥1 stage in up to 26.4% of patients), as evidenced in the MAESTRO-NASH trial. Semaglutide achieved higher rates of NASH resolution (up to 62.9%) without worsening fibrosis, especially among patients with type 2 diabetes or obesity, although fibrosis improvement was less consistently observed. Resmetirom was well tolerated with low discontinuation rates, while semaglutide was associated with more frequent, yet manageable, gastrointestinal adverse events. Conclusions: Both resmetirom and semaglutide show therapeutic potential for MASLD. Resmetirom offers more consistent antifibrotic effects, while semaglutide excels in NASH resolution and metabolic improvement. The absence of direct comparative trials underscores the need for future head-to-head studies to guide tailored treatment strategies in MASLD management. Full article
Show Figures

Figure 1

12 pages, 1434 KiB  
Article
Protective Effects of the Ethyl Acetate Fraction of Distylium racemosum Against Metabolic Dysfunction-Associated Steatohepatitis
by Young-Hyeon Lee, Min-Ho Yeo, Kyung-Soo Chang, Weon-Jong Yoon, Hye-Sook Kim, Jongwan Kim and Hye-Ran Kim
Appl. Sci. 2025, 15(13), 7238; https://doi.org/10.3390/app15137238 - 27 Jun 2025
Viewed by 309
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH), previously referred to as non-alcoholic steatohepatitis (NASH), which is a progressive non-alcoholic fatty liver disease, is accompanied by hepatic steatosis, inflammation, and fibrosis. Despite its increasing prevalence, available treatment options for MASH are limited. Here, we investigated the protective [...] Read more.
Metabolic dysfunction-associated steatohepatitis (MASH), previously referred to as non-alcoholic steatohepatitis (NASH), which is a progressive non-alcoholic fatty liver disease, is accompanied by hepatic steatosis, inflammation, and fibrosis. Despite its increasing prevalence, available treatment options for MASH are limited. Here, we investigated the protective effects of the Distylium racemosum ethyl acetate fraction (DRE) using MASH models and explored its key physiologically active components. Palmitic acid (PA)-induced AML12 hepatocytes and high-fat methionine- and choline-deficient-fed C57BL/6 mice were used as MASH models. Lipid accumulation was evaluated via triglyceride measurement, oil red O staining, and histological analysis. Lipid accumulation, inflammation, and fibrosis-associated gene expression were evaluated via real-time polymerase chain reaction. The physiologically active components of DRE were identified via high-performance liquid chromatography. Lipid accumulation and triglyceride levels were significantly reduced in PA-treated AML12 cells following DRE treatment. Additionally, DRE inhibited the expression of genes involved in lipogenesis (FAS and SREBP1c), inflammation (CD68, IL-6, and MCP-1), and fibrosis (COL1A1, COL1A2, and TIMP1). DRE reduced the liver weight, liver-to-body weight ratio, and hepatic steatosis in MASH model mice. It increased carnitine palmitoyltransferase-1 levels and decreased CD36 and transforming growth factor-β levels in the MASH mouse liver. High-performance liquid chromatography revealed that the extract contained rutin flavonoid family members. Overall, DRE was involved in lipid metabolism, inflammation, and fibrosis regulation, exerting potent hepatoprotective effects partly attributed to rutin and serving as a potential preventive candidate for MASH. Full article
Show Figures

Figure 1

23 pages, 1111 KiB  
Article
HCC in MASLD and ALD: Biochemical Pathways, Epidemiology, Diagnosis, and Treatment
by Sheel Patel, Fares Kasem, Dylan Flaherty and Ashutosh Barve
BioChem 2025, 5(3), 19; https://doi.org/10.3390/biochem5030019 - 25 Jun 2025
Viewed by 609
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality globally, with metabolic-dysfunction-associated steatohepatitis (MASH) and alcohol-related liver disease (ALD) emerging as major etiologies. This review explores the epidemiological trends, pathogenesis, and clinical management of HCC arising from MASH and ALD, highlighting both [...] Read more.
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality globally, with metabolic-dysfunction-associated steatohepatitis (MASH) and alcohol-related liver disease (ALD) emerging as major etiologies. This review explores the epidemiological trends, pathogenesis, and clinical management of HCC arising from MASH and ALD, highlighting both the shared and distinct mechanisms. MASH-HCC is driven by metabolic dysregulation, including obesity, insulin resistance, and lipotoxicity, with genetic polymorphisms such as PNPLA3 and TM6SF2 playing critical roles in disease progression. ALD-HCC, in contrast, is propelled by the toxic byproducts of ethanol metabolism, including acetaldehyde and reactive oxygen species, which induce chronic inflammation, and fibrosis. Both conditions also involve immune dysregulation, gut dysbiosis, and increased intestinal permeability, contributing to hepatic carcinogenesis. The review emphasizes that, while there is consensus regarding the screening of HCC in cirrhosis patients, there is lack of consensus on screening strategies for non-cirrhotic MASH patients who are also at risk for HCC. This underscores the importance of the early detection of cirrhosis using advanced diagnostic tools such as transient elastography and fibrosis scores. Current therapeutic approaches, ranging from surgical resection, liver transplantation, and locoregional therapies to systemic therapies like immune checkpoint inhibitors, are discussed, with an emphasis on the need for personalized treatment strategies. Finally, the review highlights future research priorities, including the development of novel biomarkers, exploration of the gut–liver axis, and deeper investigation of the interplay between genetic predisposition and environmental factors. By synthesizing these insights, the review aims to inform multidisciplinary approaches to reduce the global burden of MASH- and ALD-related HCC and improve patient outcomes. Full article
(This article belongs to the Special Issue Feature Papers in BioChem, 2nd Edition)
Show Figures

Figure 1

18 pages, 1598 KiB  
Review
Ultra-Processed Foods and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): What Is the Evidence So Far?
by Eleni V. Geladari, Dimitris Kounatidis, Gerasimos Socrates Christodoulatos, Sotiria Psallida, Argyro Pavlou, Charalampia V. Geladari, Vassilios Sevastianos, Maria Dalamaga and Natalia G. Vallianou
Nutrients 2025, 17(13), 2098; https://doi.org/10.3390/nu17132098 - 24 Jun 2025
Viewed by 953
Abstract
Ultra-processed foods (UPFs) are foods that have undergone extensive industrial processing, with the addition of emulsifiers and sweeteners together with various chemicals originating during preparation and the packaging procedures. UPFs are intended to be more palpable, long lasting, and easier to find and [...] Read more.
Ultra-processed foods (UPFs) are foods that have undergone extensive industrial processing, with the addition of emulsifiers and sweeteners together with various chemicals originating during preparation and the packaging procedures. UPFs are intended to be more palpable, long lasting, and easier to find and consume. However, their widespread use has been linked to various disorders, including insulin resistance, type 2 diabetes (T2D), obesity, and lately metabolic dysfunction-associated steatotic liver disease (MASLD). Given that MASLD is primarily driven by excessive fat accumulation in the liver and considering the high energy density and poor nutritional quality of UPFs, a plausible link has emerged between elevated UPF intake and increased MASLD risk. The aim of this review is to synthesize current data regarding the pathophysiological mechanisms underlying MASLD, the role of UPF overconsumption in its development, and potential strategies to prevent disease progression towards metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, and cirrhosis. Special focus is placed on the contribution of UPFs to these processes, highlighting the importance of minimizing their consumption as supported by contemporary research. Full article
(This article belongs to the Topic Ultra Processed Foods and Human Health)
Show Figures

Figure 1

Back to TopTop