1
|
Dai L, Liu Z, Guo C, Fan H, Zhang C, Huang J, Zhang X, Zhao S, Wang H, Zhang T. Proteomic insights into metabolic dysfunction-associated steatotic disease: Identifying therapeutic targets and assessing on-target side effects. Life Sci 2025; 373:123665. [PMID: 40287056 DOI: 10.1016/j.lfs.2025.123665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
AIMS The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rising sharply, yet treatment options remain inadequate. To uncover new therapeutic targets for MASLD, we conducted a comprehensive proteome-wide Mendelian randomization (MR) and phenome-wide association study (PheWAS). MATERIALS AND METHODS Discovery MR utilized protein quantitative trait loci (pQTL) data on 4907 plasma protein levels from 35,559 individuals, alongside genome-wide association study (GWAS) on MASLD from the Million Veteran Program (68,725 cases / 95,482 controls). Validation comprised five pairwise combinations of these discovery datasets with three additional datasets: pQTL data for 2923 proteins from the UK Biobank, and liver biopsy-confirmed MASLD GWAS (1483 cases/17,781 controls) and MRI-liver fat GWAS (31,377 subjects) (excluding discovery pair). Candidate proteins underwent druggability assessment and on-target side effect evaluation via PheWAS. KEY FINDINGS We identified 26 proteins associated with MASLD after Bonferroni correction (P < 1.16 × 10-5), with 19 of them showing no significant reverse association. Interleukin-6 (IL-6), alpha-1-antitrypsin (α1-antitrypsin), 5-hydroxytryptamine receptor 7 (5-HT7R), ephrin-B1 (EFNB1), and protein MENT (CA056) were replicated. Notably, IL-6 (OR = 2.02; 95 % CI 1.54-2.64), 5-HT7R (OR = 2.73; 95 % CI 1.96-3.80), and EFNB1 (OR = 1.82; 95 % CI 1.59-2.08) were positively associated with MASLD risk, whereas α1-antitrypsin (OR = 0.84; 95 % CI 0.78-0.90) and CA056 (OR = 0.90; 95 % CI 0.86-0.94) appeared protective. Among these, IL-6, 5-HT7R, and α1-antitrypsin were druggable. PheWAS identified potential cardiovascular side effects for 5-HT7R and α1-antitrypsin. SIGNIFICANCE The integrative study identified several plasma proteins associated with MASLD. IL-6, α1-antitrypsin, 5-HT7R, EFNB1 and CA056 deserve further investigation as potential drug targets for MASLD.
Collapse
Affiliation(s)
- Luojia Dai
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Zhenqiu Liu
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Chengnan Guo
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
| | - Hong Fan
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Chengjun Zhang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jiayi Huang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Xin Zhang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Shuzhen Zhao
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Haili Wang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, Fudan University, Shanghai 200032, China; Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Jiang J, Liu Y, Yang H, Ma Z, Liu W, Zhao M, Peng X, Qin X, Xia Y. Dietary fiber intake, genetic predisposition of gut microbiota, and the risk of metabolic dysfunction-associated steatotic liver disease. Food Res Int 2025; 211:116497. [PMID: 40356189 DOI: 10.1016/j.foodres.2025.116497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 04/16/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
This study aimed to explore the association between dietary fiber intake and the risk of metabolic dysfunction-associated steatotic liver disease (MASLD), as well as liver fat content, while considering genetic predispositions of MASLD, gut microbial abundance, and butyrate levels. This study analyzed data from 190,276 participants in the UK Biobank. Dietary fiber intake was assessed using 24-h dietary recall. MASLD cases were diagnosed through hospital admission records and death registries, and liver fat content was measured via magnetic resonance imaging. The genetic predispositions of MASLD, gut microbial abundance, and butyrate levels were evaluated using single nucleotide polymorphisms. Cox proportional hazards models were used to calculate hazard ratios (HRs) and 95 % confidence intervals (CIs). Over a median follow-up of 10.49 years, 1423 MASLD cases were recorded. Elevated dietary fiber intake was associated with a reduced risk of MASLD (HR: 0.72; 95 % CI: 0.58, 0.90) and a lower level of liver fat content (β: -0.97; 95 % CI: -1.21, -0.73) (all P for trend <0.05). Restricted cubic spline analyses further confirmed the linear inverse associations between fiber intake and the risk of MASLD. Notably, the negative associations between dietary fiber intake and both MASLD and liver fat content were consistent across different genetic predispositions of gut microbial abundance and butyrate levels. Moreover, the inverse association between dietary fiber intake and liver fat was strengthened by high genetic susceptibility of MASLD and elevated body mass index (both P for interaction <0.05). Overall, increased dietary fiber consumption was associated with a lower MASLD risk and decreased liver fat content regardless of genetic predispositions of gut microbial abundance and butyrate levels.
Collapse
Affiliation(s)
- Jinguo Jiang
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Haidian District, Beijing 100191, China.
| | - Honghao Yang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Liaoning Province, Shenyang, China.
| | - Zheng Ma
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Liaoning Province, Shenyang, China.
| | - Wenqi Liu
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| | - Maoxiang Zhao
- Interventional Center of Valvular Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing 100011, China.
| | - Xinyi Peng
- Hypertension Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Haidian District, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, China.
| | - Yang Xia
- School of Public Health, Shenyang Medical College, Shenyang, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
Valenti L, Hagström H. Bringing genetic testing into the clinical management of people with MASLD: are we there yet? J Hepatol 2025:S0168-8278(25)02247-0. [PMID: 40449686 DOI: 10.1016/j.jhep.2025.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2025] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 06/03/2025]
Affiliation(s)
- Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Precision Medicine, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Hannes Hagström
- Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Li R, Su K, Wu T, Xu L, Song W, Sun D, Zeng T, Chen J, Xin H, Li Y, Zang M, Hu M. Genome-wide enhancer-gene regulatory maps of liver reveal novel regulatory mechanisms underlying NAFLD pathogenesis. BMC Genomics 2025; 26:493. [PMID: 40375105 PMCID: PMC12082939 DOI: 10.1186/s12864-025-11668-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) represents the most widespread liver disease globally, ranging from non-alcoholic fatty liver (NAFL) and steatohepatitis (NASH) to fibrosis/cirrhosis, with potential progression to hepatocellular carcinoma (HCC). Genome-wide association studies (GWASs) have identified several single nucleotide polymorphisms (SNPs) associated with NAFLD. However, numerous GWAS signals associated with NAFLD locate in non-coding regions, posing a challenge for interpreting their functional annotation. RESULTS In this study, we utilized the Activity-by-Contact (ABC) model to construct the enhancer-gene maps of liver by integrating epigenomic data from 15 liver tissues and cell lines. We constructed the most comprehensive genome-wide regulatory maps of the liver, identifying 543,486 enhancer-gene connections, including 267,857 enhancers and 16,872 target genes. Enrichment analyses revealed that the ABC SNPs are significantly enriched in active chromatin regions and active chromatin state. By combining the ABC regulatory maps and NAFLD GWAS data, we systematically identified ABC SNPs associated with NAFLD risk. Through the functional annotations, such as pathway enrichment and drug-gene interaction analyses, we identified 6 genes (GGT1, ACTG1, SPP1, EPHA2, PROZ and SHMT1) as candidate NAFLD genes, with SHMT1 previously reported. Among the SNPs connected to the candidate genes, the ABC SNP rs2017869 (odds ratio [OR] for the C allele = 1.10, 95% CI = 1.04-1.16, P = 5.97 × 10- 4) had the highest ABC score. According to the ABC maps, rs2017869 links to GGT1, and several drugs targeting this gene, such as liothyronine, showed potential benefits to patients with NAFLD. Furthermore, we identified that another novel gene, EPHA2, may play a crucial role in NAFLD by regulating the GGT levels. CONCLUSIONS Our study provides the most comprehensive ABC regulatory maps of the liver to date. This resource offers a valuable reference for identifying regulatory variants and prioritizing susceptibility genes of liver diseases, such as NAFLD.
Collapse
Affiliation(s)
- Ruofan Li
- Medical School of Chinese People's Liberation Army (PLA), 28 Fuxing Road, 100853, Beijing, China
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Kaiyan Su
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1,838 North Guangzhou Ave, Guangzhou, Guangdong, 510515, China
| | - Tianzhun Wu
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Li Xu
- Department of Hepatopancreatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Wenyu Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Dandan Sun
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Tao Zeng
- Medical School of Chinese People's Liberation Army (PLA), 28 Fuxing Road, 100853, Beijing, China
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Jinzhang Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1,838 North Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Haibei Xin
- Department of Hepatobiliary Surgery, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Yuanfeng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| | - Mengya Zang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, 1,838 North Guangzhou Ave, Guangzhou, Guangdong, 510515, China.
| | - Minggen Hu
- Medical School of Chinese People's Liberation Army (PLA), 28 Fuxing Road, 100853, Beijing, China.
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
6
|
Byrne CD, Armandi A, Pellegrinelli V, Vidal-Puig A, Bugianesi E. Μetabolic dysfunction-associated steatotic liver disease: a condition of heterogeneous metabolic risk factors, mechanisms and comorbidities requiring holistic treatment. Nat Rev Gastroenterol Hepatol 2025; 22:314-328. [PMID: 39962331 DOI: 10.1038/s41575-025-01045-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/09/2025]
Abstract
Μetabolic dysfunction-associated steatotic liver disease (MASLD) comprises a heterogeneous condition in the presence of steatotic liver. There can be a hierarchy of metabolic risk factors contributing to the severity of metabolic dysfunction and, thereby, the associated risk of both liver and extrahepatic outcomes, but the precise ranking and combination of metabolic syndrome (MetS) traits that convey the highest risk of major adverse liver outcomes and extrahepatic disease complications remains uncertain. Insulin resistance, low-grade inflammation, atherogenic dyslipidaemia and hypertension are key to the mechanisms of liver and extrahepatic complications. The liver is pivotal in MetS progression as it regulates lipoprotein metabolism and secretes substances that affect insulin sensitivity and inflammation. MASLD affects the kidneys, heart and the vascular system, contributing to hypertension and oxidative stress. To address the global health burden of MASLD, intensified by obesity and type 2 diabetes mellitus epidemics, a holistic, multidisciplinary approach is essential. This approach should focus on both liver disease management and cardiometabolic risk factors. This Review examines the link between metabolic dysfunction and liver dysfunction and extrahepatic disease outcomes, the diverse mechanisms in MASLD due to metabolic dysfunction, and a comprehensive, personalized management model for patients with MASLD.
Collapse
Affiliation(s)
- Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vanessa Pellegrinelli
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
7
|
Ganne-Carrié N, Nahon P. Differences between hepatocellular carcinoma caused by alcohol and other aetiologies. J Hepatol 2025; 82:909-917. [PMID: 39710147 DOI: 10.1016/j.jhep.2024.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/14/2024] [Accepted: 12/07/2024] [Indexed: 12/24/2024]
Abstract
Alcohol-related liver disease is the third leading cause of hepatocellular carcinoma worldwide and the leading cause in Europe. Additionally, the recent definition of metabolic dysfunction-associated steatotic liver disease with increased alcohol intake (MetALD) will enrich this population with a more nuanced phenotype, reflecting recent epidemiological trends. In these patients, the hepatocellular carcinoma diagnosis is often delayed and less frequently detected through screening programmes. Moreover, at the time of diagnosis, patients with alcohol-related hepatocellular carcinoma tend to have a poorer general condition, more severely impaired liver function, and a higher prevalence of comorbidities, leading to increased competitive mortality. However, when hepatocellular carcinoma is diagnosed during surveillance programmes in patients with alcohol-related liver disease or MetALD, the rate of allocation to first-line curative treatments is high (56%) and comparable to that of patients with virus-related hepatocellular carcinoma. As a consequence, the aetiology of the underlying cirrhosis cannot be considered an independent prognostic factor in patients with hepatocellular carcinoma. Instead, prognosis is driven by liver function, general condition, and tumour burden. This underscores the crucial role of early diagnosis through periodic surveillance in patients with alcohol- or MetALD-related cirrhosis.
Collapse
Affiliation(s)
- Nathalie Ganne-Carrié
- AP-HP, Hôpital Avicenne, Liver Unit, F-93000 Bobigny, France; University Sorbonne Paris Nord, UFR SMBH, F-93000 Bobigny, France; INSERM UMR-1168, Functional Genomics of Solid Tumours, F-75006 Paris, France.
| | - Pierre Nahon
- AP-HP, Hôpital Avicenne, Liver Unit, F-93000 Bobigny, France; University Sorbonne Paris Nord, UFR SMBH, F-93000 Bobigny, France; INSERM UMR-1168, Functional Genomics of Solid Tumours, F-75006 Paris, France
| |
Collapse
|
8
|
Wang J, Mo J, Wan X, Fan Y, Zhuang P. Meat and fish consumption, genetic risk and risk of severe metabolic-associated fatty liver disease: a prospective cohort of 487,875 individuals. Nutr J 2025; 24:65. [PMID: 40281620 PMCID: PMC12023461 DOI: 10.1186/s12937-025-01134-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Diet, specifically meat consumption, has been implicated as a modifiable risk factor in the development of metabolic-associated fatty liver disease (MAFLD). This study aimed to investigate the associations between various types of meat intake and the risk of severe MAFLD and to examine whether genetic risk influences these associations. METHODS This research utilized data from the UK Biobank, which initially enrolled over 500,000 participants between 2006 and 2010, of whom 487,875 were eligible for our analyses. Meat intake, including unprocessed red meat, processed meat, poultry, and fish, was evaluated through a validated touchscreen questionnaire. Cox proportional hazards models were used to analyze the relationship between meat consumption and severe MAFLD risk, adjusting for potential confounders. Genetic risk scores (GRS) were calculated using five MAFLD-associated SNPs, allowing for analyses of gene-diet interactions. RESULTS During a follow-up period totaling 6,036,554 person-years (mean duration: 12.1 years), 5,731 new cases of severe MAFLD were identified. High intakes of total meat, processed meat, unprocessed red meat and poultry were associated with increased MAFLD risk, with adjusted hazard ratios (HR) of 1.76 (95% CI: 1.33-2.33), 1.19 (1.02-1.40), 1.34 (1.17-1.53), and 1.21 (0.98-1.49), respectively, for the highest versus lowest intake categories. In contrast, oily fish intake showed a protective association (HR: 0.72; 95% CI: 0.53-0.97). No significant interaction was observed between meat intake and GRS for any meat subtype, suggesting that the associations were independent of genetic predisposition. CONCLUSIONS High consumption of red and processed meat was associated with an increased risk of severe MAFLD, while oily fish intake showed an inverse association with the risk of MAFLD. These effects were consistent across genetic risk levels for MAFLD. Our findings reinforce dietary recommendations to limit red and processed meat and encourage oily fish intake for MAFLD prevention, irrespective of individual genetic risk.
Collapse
Affiliation(s)
- Jianjin Wang
- Department of Clinical Medicine, Shaoxing University School of Medicine, Zhejiang, 312000, Shaoxing, China
| | - Jianshu Mo
- Department of Secondary Internal Medicine, Yuyao Hospital of Traditional Chinese Medicine, Yuyao, 315400, Zhejiang, China
| | - Xuzhi Wan
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Yilei Fan
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Zhejiang Police College, Hangzhou, 310053, Zhejiang, China.
| | - Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China.
| |
Collapse
|
9
|
Liukkonen V, Semenova M, Hyvärinen K, Lauronen J, Partanen J, Arola J, Nordin A, Färkkilä M, Åberg F. Genetic Risk Factors for Steatotic Liver Disease After Liver Transplantation. Liver Int 2025; 45:e70067. [PMID: 40087975 DOI: 10.1111/liv.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND The role of genetic risk factors for steatotic liver disease (SLD) is intriguing in liver transplantation (LT), as both donor and recipient genetic factors may play a role. There are only a few small-scale studies published so far. METHODS We analysed the incidence and risk factors for post-LT SLD and the impact of 56 SLD-associated genetic variants in 595 donor-recipient pairs with liver biopsy available ≥ 6 months after LT. We evaluated whether the polygenic risk score (PRS-5) improves the ability to predict post-LT SLD in addition to non-genetic risk factors. RESULTS SLD after LT was diagnosed in 34.5% of patients during a median 7.6-year follow-up. In multivariate analysis including non-genetic risk factors, donor PNPLA3 rs738409-G (HR for SLD: C/G 1.34, p = 0.051, G/G 2.25, p = 0.004), donor HSD17B13 rs72613567-TA (HR for SLD: TA/T 0.68, p = 0.02 TA/TA HR 0.50, p = 0.10) and recipient UCP2 rs695366-G (HR for SLD: A/G 0.63, p = 0.002, G/G HR 0.50, p = 0.04) appeared as the most important genetic risk factors for post-LT SLD. The addition of PRS-5 to a multivariate regression model (including non-genetic risk factors) improved the predictive ability for SLD only modestly (AUC 0.78 to 0.80). CONCLUSIONS Various genetic variants contribute to post-LT SLD with separate variants among recipients and donors, with donor PNPLA3 rs738409-G as the most significant risk allele. Still, donor and recipient genotyping provide only modest additional value for individual risk stratification over phenotype data, highlighting the role of modifiable risk factors.
Collapse
Affiliation(s)
- Ville Liukkonen
- North Karelia Central Hospital, Gastroenterology, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| | - Maria Semenova
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Kati Hyvärinen
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Jouni Lauronen
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Jukka Partanen
- Finnish Red Cross Blood Service, Research & Development, Helsinki, Finland
| | - Johanna Arola
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pathology, Helsinki University Hospital, Diagnostic Centre, Helsinki, Finland
| | - Arno Nordin
- Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Martti Färkkilä
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Fredrik Åberg
- Transplantation and Liver Surgery, Helsinki University Hospital, University of Helsinki, Faculty of Medicine, Helsinki, Finland
| |
Collapse
|
10
|
Di Sessa A, Zarrilli S, Forcina G, Frattolillo V, Camponesco O, Migliaccio C, Ferrara S, Umano GR, Cirillo G, Miraglia Del Giudice E, Marzuillo P. Role of metabolic dysfunction-associated steatotic liver disease and of its genetics on kidney function in childhood obesity. Int J Obes (Lond) 2025; 49:605-611. [PMID: 39521922 DOI: 10.1038/s41366-024-01674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Evidence linked metabolic associated steatotic liver disease (MASLD) to kidney damage with the potential contribution of the I148M variant of the Patatin-like phospholipase containing domain 3 (PNPLA3) gene. We aimed at investigating the relationship of MASLD and of its genetics with kidney function in children with obesity. METHODS A comprehensive evaluation including genotyping for the I148M PNPLA3 polymorphism was performed in 1037 children with obesity. Fatty liver (FL) was assessed by liver ultrasound. According to MASLD criteria, subjects with obesity but without FL were included in group 1, while patients with obesity and FL (encompassing one MASLD criterion) were clustered into group 2. Group 3 included patients with obesity, FL, and metabolic dysregulation (encompassing >1 MASLD criterion). RESULTS Alanine transaminase levels significantly increased while estimated glomerular filtration rate (eGFR) significantly reduced from group 1 to 3. Group 3 showed a higher percentage of carriers of the I148M allele of the PNPLA3 gene compared to other groups (p < 0.0001). Carriers of group 2 and of group 3 showed reduced eGFR levels than noncarriers of group 2 (p = 0.04) and of group 3 (p = 0.02), respectively. A general linear model for eGFR variance in the study population showed an inverse association of eGFR with both MASLD and PNPLA3 genotypes (p = 0.011 and p = 0.02, respectively). An inverse association of eGFR with MASLD was also confirmed only in carriers (p = 0.006). CONCLUSIONS The coexistence of more than 1 MASLD criterion in children with obesity seems to adversely affect kidney function. The PNPLA3 I148M allele further impacts on this association.
Collapse
Affiliation(s)
- Anna Di Sessa
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Sarah Zarrilli
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianmario Forcina
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vittoria Frattolillo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ornella Camponesco
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudia Migliaccio
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Serena Ferrara
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppina Rosaria Umano
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Grazia Cirillo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Emanuele Miraglia Del Giudice
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
11
|
Lu MY, Wei YJ, Wang CW, Liang PC, Yeh ML, Tsai YS, Tsai PC, Ko YM, Lin CC, Chen KY, Lin YH, Jang TY, Hsieh MY, Lin ZY, Huang CF, Huang JF, Dai CY, Chuang WL, Yu ML. Mitochondrial mt12361A>G increased risk of metabolic dysfunction-associated steatotic liver disease among non-diabetes. World J Gastroenterol 2025; 31:103716. [PMID: 40093674 PMCID: PMC11886537 DOI: 10.3748/wjg.v31.i10.103716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/16/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Insulin resistance, lipotoxicity, and mitochondrial dysfunction contribute to the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). Mitochondrial dysfunction impairs oxidative phosphorylation and increases reactive oxygen species production, leading to steatohepatitis and hepatic fibrosis. Artificial intelligence (AI) is a potent tool for disease diagnosis and risk stratification. AIM To investigate mitochondrial DNA polymorphisms in susceptibility to MASLD and establish an AI model for MASLD screening. METHODS Multiplex polymerase chain reaction was performed to comprehensively genotype 82 mitochondrial DNA variants in the screening dataset (n = 264). The significant mitochondrial single nucleotide polymorphism was validated in an independent cohort (n = 1046) using the Taqman® allelic discrimination assay. Random forest, eXtreme gradient boosting, Naive Bayes, and logistic regression algorithms were employed to construct an AI model for MASLD. RESULTS In the screening dataset, only mt12361A>G was significantly associated with MASLD. mt12361A>G showed borderline significance in MASLD patients with 2-3 cardiometabolic traits compared with controls in the validation dataset (P = 0.055). Multivariate regression analysis confirmed that mt12361A>G was an independent risk factor of MASLD [odds ratio (OR) = 2.54, 95% confidence interval (CI): 1.19-5.43, P = 0.016]. The genetic effect of mt12361A>G was significant in the non-diabetic group but not in the diabetic group. mt12361G carriers had a 2.8-fold higher risk than A carriers in the non-diabetic group (OR = 2.80, 95%CI: 1.22-6.41, P = 0.015). By integrating clinical features and mt12361A>G, random forest outperformed other algorithms in detecting MASLD [training area under the receiver operating characteristic curve (AUROC) = 1.000, validation AUROC = 0.876]. CONCLUSION The mt12361A>G variant increased the severity of MASLD in non-diabetic patients. AI supports the screening and management of MASLD in primary care settings.
Collapse
Affiliation(s)
- Ming-Ying Lu
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung 80708, Taiwan
| | - Yu-Ju Wei
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chih-Wen Wang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Po-Cheng Liang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Ming-Lun Yeh
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Yi-Shan Tsai
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Pei-Chien Tsai
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Yu-Min Ko
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Ching-Chih Lin
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Kuan-Yu Chen
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Yi-Hung Lin
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Tyng-Yuan Jang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Ming-Yen Hsieh
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Zu-Yau Lin
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chung-Feng Huang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Jee-Fu Huang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chia-Yen Dai
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Wan-Long Chuang
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Ming-Lung Yu
- Hepatitis Center and Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung 80708, Taiwan
| |
Collapse
|
12
|
Cherubini A, Rosso C, Della Torre S. Sex-specific effects of PNPLA3 I148M. Liver Int 2025; 45:e16088. [PMID: 39262132 PMCID: PMC11815604 DOI: 10.1111/liv.16088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD, previously termed NAFLD, nonalcoholic fatty liver disease) is a complex multifactorial disease showing generally higher prevalence and severity in men than in women. With respect to women, men are also more prone to develop metabolic dysfunction-associated steatohepatitis, fibrosis and liver-related complications. Several genetic, hormonal, environmental and lifestyle factors may contribute to sex differences in MASLD development, progression and outcomes. However, after menopause, the sex-specific prevalence of MASLD shows an opposite trend between men and women, pointing to the relevance of oestrogen signalling in the sexual dimorphism of MASLD. The patatin-like phospholipase domain-containing protein 3 (PNPLA3) gene, that encodes a triacylglycerol lipase that plays a crucial role in lipid metabolism, has emerged as a key player in the pathogenesis of MASLD, with the I148M variant being strongly associated with increased liver fat content and disease severity. Recent advances indicate that carrying the PNPLA3 I148M variant can be a risk factor for MASLD especially for women. To elucidate the molecular mechanisms underlying the sex-specific role of PNPLA3 I148M in the development of MASLD, several in vitro, ex vivo and in vivo models have been developed.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Department of Transfusion Medicine, Precision Medicine—Biological Resource CenterFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Chiara Rosso
- Department of Medical SciencesUniversity of TurinTurinItaly
| | - Sara Della Torre
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
13
|
Thiele M, Johansen S, Israelsen M, Trebicka J, Abraldes JG, Gines P, Krag A. Noninvasive assessment of hepatic decompensation. Hepatology 2025; 81:1019-1037. [PMID: 37801593 PMCID: PMC11825506 DOI: 10.1097/hep.0000000000000618] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 07/19/2023] [Indexed: 10/08/2023]
Abstract
Noninvasive tests (NITs) are used in all aspects of liver disease management. Their most prominent break-through since the millennium has been in advancing early detection of liver fibrosis, but their use is not limited to this. In contrast to the symptom-driven assessment of decompensation in patients with cirrhosis, NITs provide not only opportunities for earlier diagnoses but also accurate prognostication, targeted treatment decisions, and a means of monitoring disease. NITs can inform disease management and decision-making based on validated cutoffs and standardized interpretations as a valuable supplement to clinical acumen. The Baveno VI and VII consensus meetings resulted in tangible improvements to pathways of care for patients with compensated and decompensated advanced chronic liver disease, including the combination of platelet count and transient elastography to diagnose clinically significant portal hypertension. Furthermore, circulating NITs will play increasingly important roles in assessing the response to interventions against ascites, variceal bleeding, HE, acute kidney injury, and infections. However, due to NITs' wide availability, there is a risk of inaccurate use, leading to a waste of resources and flawed decisions. In this review, we describe the uses and pitfalls of NITs for hepatic decompensation, from risk stratification in primary care to treatment decisions in outpatient clinics, as well as for the in-hospital management of patients with acute-on-chronic liver failure. We summarize which NITs to use when, for what indications, and how to maximize the potential of NITs for improved patient management.
Collapse
Affiliation(s)
- Maja Thiele
- Department of Gastroenterology and Hepatology, Fibrosis, Fatty Liver and Steatohepatitis Research Center Odense (FLASH), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Stine Johansen
- Department of Gastroenterology and Hepatology, Fibrosis, Fatty Liver and Steatohepatitis Research Center Odense (FLASH), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Mads Israelsen
- Department of Gastroenterology and Hepatology, Fibrosis, Fatty Liver and Steatohepatitis Research Center Odense (FLASH), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Jonel Trebicka
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Internal Medicine B, University of Münster, Münster, Germany
- European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
| | - Juan G. Abraldes
- Division of Gastroenterology, University of Alberta, Edmonton, Canada
| | - Pere Gines
- Liver Unit, Hospital Clínic of Barcelona, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Spain
- Institute of Biomedical Investigation August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEReHD), Barcelona, Spain
| | - Aleksander Krag
- Department of Gastroenterology and Hepatology, Fibrosis, Fatty Liver and Steatohepatitis Research Center Odense (FLASH), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
14
|
Bourganou MV, Chondrogianni ME, Kyrou I, Flessa CM, Chatzigeorgiou A, Oikonomou E, Lambadiari V, Randeva HS, Kassi E. Unraveling Metabolic Dysfunction-Associated Steatotic Liver Disease Through the Use of Omics Technologies. Int J Mol Sci 2025; 26:1589. [PMID: 40004054 PMCID: PMC11855544 DOI: 10.3390/ijms26041589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is the most prevalent liver disorder globally, linked to obesity, type 2 diabetes, and cardiovascular risk. Understanding its potential progression from simple steatosis to cirrhosis and hepatocellular carcinoma (HCC) is crucial for patient management and treatment strategies. The disease's complexity requires innovative approaches for early detection and personalized care. Omics technologies-such as genomics, transcriptomics, proteomics, metabolomics, and exposomics-are revolutionizing the study of MASLD. These high-throughput techniques allow for a deeper exploration of the molecular mechanisms driving disease progression. Genomics can identify genetic predispositions, whilst transcriptomics and proteomics reveal changes in gene expression and protein profiles during disease evolution. Metabolomics offers insights into the metabolic alterations associated with MASLD, while exposomics links environmental exposures to MASLD progression and pathology. By integrating data from various omics platforms, researchers can map out the intricate biochemical pathways involved in liver disease progression. This review discusses the roles of omics technologies in enhancing the understanding of disease progression and highlights potential diagnostic and therapeutic targets within the MASLD spectrum, emphasizing the need for non-invasive tools in disease staging and treatment development.
Collapse
Affiliation(s)
- Maria V. Bourganou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 IGB, UK
| | - Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaia Lambadiari
- 2nd Department of Internal-Medicine, Diabetes Centre, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
15
|
Ananchuensook P, Moonlisarn K, Boonkaew B, Bunchorntavakul C, Tangkijvanich P. Diagnostic Performance of Serum Mac-2-Binding Protein Glycosylation Isomer as a Fibrosis Biomarker in Non-Obese and Obese Patients with MASLD. Biomedicines 2025; 13:415. [PMID: 40002828 PMCID: PMC11853689 DOI: 10.3390/biomedicines13020415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Serum mac-2-binding protein glycosylation isomer (M2BPGi) is a new biomarker for liver fibrosis. However, its performance in metabolic dysfunction-associated steatotic liver disease (MASLD), particularly in obese patients, remains to be explored. Methods: This study evaluated the role of M2BPGi in predicting liver fibrosis in 205 patients with MASLD using magnetic resonance elastography (MRE) as a reference. The performance of M2BPGi was compared to vibration-controlled transient elastography (VCTE), FIB-4, APRI, and NFS. The PNPLA3, TM6SF2, and HSD17B13 polymorphisms were assessed by allelic discrimination assays. Results: The area under the ROC curves for VCTE, M2BPGi FIB-4, APRI, and NFS in differentiating significant fibrosis were 0.95 (95% CI; 0.91-0.98), 0.85 (0.79-0.92), 0.81 (0.74-0.89), 0.79 (0.71-0.87), and 0.80 (0.72-0.87) (all p < 0.001), respectively. The optimal cut-off values of M2BPGi in predicting significant fibrosis, advanced fibrosis, and cirrhosis were 0.82, 0.95, and 1.23 cut-off index (COI); yielding satisfactory sensitivity, specificity, and diagnostic accuracy. The performance of M2BPGi was consistent among subgroups according to BMI, while the AUROCs of FIB-4, APRI, and NFS were remarkably decreased in patients with BMI ≥ 30 kg/m2. Patients with the PNPLA3 GG genotype had significantly higher M2BPGi than those with the CC/CG genotypes. In multivariate analysis, the independent factors associated with significant liver fibrosis were VCTE, M2BPGi, and PNPLA3 rs738409. Conclusions: Our data demonstrated that serum M2BPGi accurately assessed liver fibrosis across different BMI, indicating that this biomarker could apply to non-obese and obese patients with MASLD in clinical settings.
Collapse
Affiliation(s)
- Prooksa Ananchuensook
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Academic Affair, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kamonchanok Moonlisarn
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (B.B.)
| | - Bootsakorn Boonkaew
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (B.B.)
| | | | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (B.B.)
| |
Collapse
|
16
|
Liu Z, Chen X, Yuan H, Jin L, Zhang T, Chen X. Dissecting the shared genetic architecture between nonalcoholic fatty liver disease and type 2 diabetes. Hum Mol Genet 2025; 34:338-346. [PMID: 39690818 DOI: 10.1093/hmg/ddae184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Observational studies have reported a bidirectional correlation between nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), but the shared genetic basis between the two conditions remains unclear. Using genome-wide association study (GWAS) summary data from European-ancestry populations, we examined the cross-trait genetic correlation and identified genomic overlaps and shared risk loci. We employed a latent causal variable model and Mendelian randomization (MR) analysis to infer causal relationships. Colocalization analysis and conditional/conjunctional false discovery rate (condFDR/conjFDR) were used to identify genomic overlaps and shared risk loci. Two-step MR analysis was utilized to identify potential mediators. We observed a strong positive genomic correlation between NAFLD and T2D (rg = 0.652, P = 5.67 × 10-6) and identified tissue-specific transcriptomic correlations in the pancreas, liver, skeletal muscle, subcutaneous adipose, and blood. Genetic enrichment was observed in NAFLD conditional on associations with T2D and vice versa, indicating significant polygenic overlaps. We found robust evidence for the causal effect of NAFLD on T2D, particularly insulin-related T2D, rather than vice versa. Colocalization analysis identified shared genomic regions between NAFLD and T2D, including GCKR, FTO, MAU2-TM6SF2, and PNPLA3-SAMM50. High-density lipoprotein cholesterol and insulin were partly mediated the association between NAFLD and T2D. These findings unveil a close genetic link between NAFLD and T2D, shedding light on the biological mechanisms connecting NAFLD progression to T2D.
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Xiaochen Chen
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South RD, Shanghai 200025, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, 130 Dong'an RD, Shanghai 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, 130 Dong'an RD, Shanghai 200032, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Urumqi RD, Shanghai 200040, China
| |
Collapse
|
17
|
Caddeo A, Romeo S. Precision medicine and nucleotide-based therapeutics to treat steatotic liver disease. Clin Mol Hepatol 2025; 31:S76-S93. [PMID: 39103998 PMCID: PMC11925435 DOI: 10.3350/cmh.2024.0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/07/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex multifactorial disease and becoming the leading cause of liver-related morbidity and mortality. MASLD spans from isolated steatosis to metabolic dysfunction-associated steatohepatitis (MASH), that may progress to cirrhosis and hepatocellular carcinoma (HCC). Genetic, metabolic, and environmental factors strongly contribute to the heterogeneity of MASLD. Lifestyle intervention and weight loss represent a viable treatment for MASLD. Moreover, Resmetirom, a thyroid hormone beta receptor agonist, has recently been approved for MASLD treatment. However, most individuals treated did not respond to this therapeutic, suggesting the need for a more tailored approach to treat MASLD. Oligonucleotide-based therapies, namely small-interfering RNA (siRNA) and antisense oligonucleotide (ASO), have been recently developed to tackle MASLD by reducing the expression of genes influencing MASH progression, such as PNPLA3 and HSD17B13. Here, we review the latest progress made in the synthesis and development of oligonucleotide-based agents targeting genetic determinants of MASH.
Collapse
Affiliation(s)
- Andrea Caddeo
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Stefano Romeo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Medicine, Endocrinology (H7) Karolinska Institute and Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
18
|
Bhardwaj M, Mazumder PM. An insight on the additive impact of type 2 diabetes mellitus and nonalcoholic fatty liver disease on cardiovascular consequences. Mol Biol Rep 2025; 52:169. [PMID: 39873861 DOI: 10.1007/s11033-025-10249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) are associated with a multifactorial complicated aetiology that is often coexisting and has a strong and distinct connection with cardiovascular diseases (CVDs). In order to accomplish effective and appropriate therapeutic strategies, a deeper understanding of the bidirectional interaction between NAFLD patients, NAFLD patients with T2DM, and NAFLD patients with CVDs is required to control the concomitant rise in prevalence of these conditions worldwide. This article also aims to shed light on the epidemiology and mechanisms behind the relationship between T2DM, NAFLD and the related cardiovascular consequences. METHOD Literature was collected from PubMed, Medline, Embase, Web of Science and Google scholar from inception to June, 2024. For surveying literature different combinations and formats of terms including NAFLD, NASH, T2DM and CVDs were used. RESULTS In the recent decade, clinical and epidemiological studies have been conducted and provide strong evidence that NAFLD is closely linked with CVD progression along with associated morbidity and mortality in both patients with and without T2DM. Several mechanistic approaches contribute to cardiovascular consequences and abnormalities in cardiac biomarkers in T2DM and NAFLD patients, including adipose tissue malfunction, mitochondrial dysfunction, the microbiota, genetic and epigenetic alterations contributing to insulin resistance, glucotoxicity and lipotoxicity. CONCLUSION The study reveals a complex interplay between diabetes, hepatic and cardiovascular complications, leading to significant morbidity and mortality in diabetic and NAFLD patients. This pandemic necessitates further research to identify mitigating variables and develop effective treatment approaches.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India.
| |
Collapse
|
19
|
Dileo E, Saba F, Parasiliti-Caprino M, Rosso C, Bugianesi E. Impact of Sexual Dimorphism on Therapy Response in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease: From Conventional and Nutritional Approaches to Emerging Therapies. Nutrients 2025; 17:477. [PMID: 39940335 PMCID: PMC11821005 DOI: 10.3390/nu17030477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a spectrum of liver disease ranging from hepatic fat accumulation to steatohepatitis (metabolic dysfunction-associated steatohepatitis, MASH), fibrosis, cirrhosis, and potentially hepatocellular carcinoma in the absence of excessive alcohol consumption. MASLD is characterized by substantial inter-individual variability in terms of severity and rate of progression, with a prevalence that is generally higher in men than in women. Steroids metabolism is characterized by sexual dimorphism and may have an impact on liver disease progression; indeed, several therapeutic strategies targeting hormone receptors are under phase 2/3 development. Despite the fact that the importance of sexual dimorphism in the setting of MASLD is well recognized, the underlying molecular mechanisms that can potentially drive the disease toward progression are not clear. The aim of this review is to delve into the crosstalk between sexual dimorphism and steroid hormone perturbation under nutritional and pharmacological intervention.
Collapse
Affiliation(s)
| | | | | | - Chiara Rosso
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.D.); (F.S.); (M.P.-C.)
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.D.); (F.S.); (M.P.-C.)
| |
Collapse
|
20
|
Porada M, Bułdak Ł. From Pathophysiology to Practice: Evolving Pharmacological Therapies, Clinical Complications, and Pharmacogenetic Considerations in Portal Hypertension. Metabolites 2025; 15:72. [PMID: 39997697 PMCID: PMC11857179 DOI: 10.3390/metabo15020072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Portal hypertension is a major complication of chronic liver diseases, leading to serious issues such as esophageal variceal bleeding. The increase in portal vein pressure is driven by both an organic component and a functional component, including tonic contraction of hepatic stellate cells. These processes result in a pathological rise in intrahepatic vascular resistance, stemming from partial impairment of hepatic microcirculation, which is further exacerbated by abnormalities in extrahepatic vessels, including increased portal blood flow. Objectives: This review aims to provide a comprehensive overview of the evolving pharmacological therapies for portal hypertension, with consideration and discussion of pathophysiological mechanisms, clinical complications, and pharmacogenetic considerations, highlighting potential directions for future research. Methods: A review of recent literature was performed to evaluate current knowledge and potential therapeutic strategies in portal hypertension. Results: For over 35 years, non-selective beta-blockers have been the cornerstone therapy for portal hypertension by reducing portal vein inflow as an extrahepatic target, effectively preventing decompensation and variceal hemorrhages. However, since not all patients exhibit an adequate response to non-selective beta-blockers (NSBBs), and some may not tolerate NSBBs, alternative or adjunctive therapies that enhance the effects of NSBBs on portal pressure are being investigated in preclinical and early clinical studies. Conclusions: A better understanding of pharmacogenetic factors and pathophysiological mechanisms could lead to more individualized and effective treatments for portal hypertension. These insights highlight potential directions for future research.
Collapse
Affiliation(s)
- Michał Porada
- Students’ Scientific Society, Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland;
| | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
| |
Collapse
|
21
|
Galvanin C, Buch S, Nahon P, Trépo E. PNPLA3 in Alcohol-Related Liver Disease. Liver Int 2025; 45:e16211. [PMID: 39679853 DOI: 10.1111/liv.16211] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
The discovery of PNPLA3 as a genetic risk factor for liver disease has transformed our understanding of the pathogenesis of alcohol-related liver disease (ALD). The recent reclassification of fatty liver disease as steatotic liver disease (SLD), introducing metabolic dysfunction and alcohol-related liver disease (MetALD), has highlighted how genetic and environmental factors synergistically drive liver damage. The PNPLA3 rs738409 variant stands as a paradigmatic example of gene-environment interaction, where its effect on liver disease is dramatically amplified by alcohol consumption, obesity and type 2 diabetes. Understanding these interactions has revealed novel pathogenic mechanisms. The robust genetic evidence and a growing understanding of molecular mechanisms have made PNPLA3 an attractive therapeutic target. Several compounds targeting PNPLA3 are now in clinical development. While initial trials have focused on metabolic dysfunction-associated SLD, the recognition that almost all individuals with excessive alcohol consumption have metabolic comorbidities provides an unprecedented opportunity to evaluate these genetically informed therapies in MetALD. In this review, we examine the role of PNPLA3 in ALD, focusing on gene-environment interactions and therapeutic implications in the context of the new disease classification framework.
Collapse
Affiliation(s)
- Clélia Galvanin
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Stephan Buch
- Department of Medicine I, Dresden University Hospital, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Pierre Nahon
- Service d'Hépatologie, AP-HP Avicenne, Bobigny, France
- Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Team «Functional Genomics of Solid Tumors», Paris, France
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
22
|
Wei P, Chen K, Chen J. Engineering an Ultrasound-Responsive Glycopolymersome for Hepatocyte-Specific Gene Delivery. Biomacromolecules 2024; 25:7838-7849. [PMID: 39514216 DOI: 10.1021/acs.biomac.4c01128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The ability to design liver-targeted gene delivery vectors is plagued with difficulties ranging from carrier-mediated cellular toxicity to challenges in encapsulating sensitive nucleic acids. Herein, we present an ultrasound-responsive glycopolymersome strategy for in situ loading of nucleic acids and achieving hepatocyte-specific gene delivery. This glycopolymersome is self-assembled from a block copolymer, N-acetylgalactosamine-grafted poly(glutamic acid)-block-poly(ε-caprolactone) (PGAGalNAc-b-PCL). GalNAc is introduced to afford liver targeting through the selective binding to the asialoglycoprotein receptor overexpressed on hepatocytes. External ultrasound is utilized to assist in encapsulating nucleic acids within the hydrophilic lumen of glycopolymersomes by exploiting their ultrasound responsiveness nature. Biological studies confirmed the successful encapsulation of plasmid DNA (pDNA) and small interfering RNA (siRNA), rapid nuclear internalization, and efficient gene transfection. These findings collectively demonstrated that this ultrasound-responsive glycopolymersome could be exploited as a novel safe and efficient gene vector targeting hepatocytes.
Collapse
Affiliation(s)
- Ping Wei
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Kai Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
23
|
La Mura V, Colombo M, Foster GR, Angeli P, Miesbach W, Klamroth R, Pierce GF, O'Mahony B, Lim MY, Hernandez-Gea V, Makris M, Peyvandi F. The management of liver disease in people with congenital bleeding disorders: guidance from European Association for Haemophilia and Allied Disorders, European Haemophilia Consortium, ISTH, and World Federation of Hemophilia. J Thromb Haemost 2024; 22:3629-3639. [PMID: 39271019 DOI: 10.1016/j.jtha.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
People with bleeding disorders (PWBD) have been exposed to the risk of developing chronic viral hepatitis and cirrhosis after replacement therapy. Today, the advent of new pharmacologic strategies for the control of hemostasis and the efficacious antiviral therapies against hepatitis C virus and hepatitis B virus have significantly reduced this risk. However, the definitive success for liver health in this clinical setting is also influenced by other factors, such as the severity of liver disease at the time of hepatitis B virus/hepatitis C virus antiviral therapy and the exposure to highly prevalent factors of chronic liver damage (eg, metabolic dysfunction and/or alcohol) that can cause a residual risk of complications such as hepatocellular carcinoma, portal hypertension, and liver insufficiency. With this background, a group of experts selected among hepatologists, hematologists, PWBD treaters, and patient representatives produced this practical multisociety guidance for the protection of liver health and the prevention and management of liver complications in PWBD based on the most updated protocols of care.
Collapse
Affiliation(s)
- Vincenzo La Mura
- Fondazione Istituto Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi and Thrombosis Center, Milan, Italy; Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Massimo Colombo
- European Association for the Study of the Liver (EASL) International Liver Foundation, Geneva, Switzerland
| | - Graham R Foster
- Barts Liver Centre, Blizard Institute, Queen Mary University of London (QMUL), London, United Kingdom
| | - Paolo Angeli
- Unit of Internal Medicine and Hepatology (UIMH), Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Wolfgang Miesbach
- Department of Haemostasis/Haemophilia Centre, Medical Clinic 2, University Hospital Frankfurt, Frankfurt, Germany
| | - Robert Klamroth
- Department for Internal Medicine and Vascular Medicine, Haemophilia Treatment Center, Vivantes Hospital im Friedrichshain, Berlin, Germany
| | - Glenn F Pierce
- World Federation of Hemophilia (WFH), Montreal, Quebec, Canada
| | - Brian O'Mahony
- Irish Haemophilia Society, Dublin, Ireland; Trinity College Dublin, Dublin, Ireland
| | - Ming Y Lim
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Virginia Hernandez-Gea
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Care Provider of the European Reference Network on Rare Liver Disorders (ERN-RareLiver), Universitat de Barcelona, Barcelona, Spain
| | - Michael Makris
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Flora Peyvandi
- Fondazione Istituto Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi and Thrombosis Center, Milan, Italy; Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.
| |
Collapse
|
24
|
Burra P, Zanetto A, Schnabl B, Reiberger T, Montano-Loza AJ, Asselta R, Karlsen TH, Tacke F. Hepatic immune regulation and sex disparities. Nat Rev Gastroenterol Hepatol 2024; 21:869-884. [PMID: 39237606 DOI: 10.1038/s41575-024-00974-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/07/2024]
Abstract
Chronic liver disease is a major cause of morbidity and mortality worldwide. Epidemiology, clinical phenotype and response to therapies for gastrointestinal and liver diseases are commonly different between women and men due to sex-specific hormonal, genetic and immune-related factors. The hepatic immune system has unique regulatory functions that promote the induction of intrahepatic tolerance, which is key for maintaining liver health and homeostasis. In liver diseases, hepatic immune alterations are increasingly recognized as a main cofactor responsible for the development and progression of chronic liver injury and fibrosis. In this Review, we discuss the basic mechanisms of sex disparity in hepatic immune regulation and how these mechanisms influence and modify the development of autoimmune liver diseases, genetic liver diseases, portal hypertension and inflammation in chronic liver disease. Alterations in gut microbiota and their crosstalk with the hepatic immune system might affect the progression of liver disease in a sex-specific manner, creating potential opportunities for novel diagnostic and therapeutic approaches to be evaluated in clinical trials. Finally, we identify and propose areas for future basic, translational and clinical research that will advance our understanding of sex disparities in hepatic immunity and liver disease.
Collapse
Affiliation(s)
- Patrizia Burra
- Gastroenterology and Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padua University Hospital, Padua, Italy.
| | - Alberto Zanetto
- Gastroenterology and Multivisceral Transplant Unit, Department of Surgery, Oncology, and Gastroenterology, Padua University Hospital, Padua, Italy
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Aldo J Montano-Loza
- Division of Gastroenterology and Liver Unit, Department of Medicine, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Tom Hemming Karlsen
- Department of Transplantation Medicine, Clinic of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital and University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Clinic of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| |
Collapse
|
25
|
Ahmed A, Cule M, Bell JD, Sattar N, Yaghootkar H. Differing genetic variants associated with liver fat and their contrasting relationships with cardiovascular diseases and cancer. J Hepatol 2024; 81:921-929. [PMID: 38960375 DOI: 10.1016/j.jhep.2024.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND & AIMS The mechanisms underlying the association of steatotic liver disease with cardiovascular and cancer outcomes are poorly understood. We aimed to use MRI-derived measures of liver fat and genetics to investigate causal mechanisms that link higher liver fat to various health outcomes. METHODS We conducted a genome-wide association study on 37,358 UK Biobank participants to identify genetic variants associated with liver fat measured from MRI scans. We used a Mendelian randomisation approach to investigate the causal effect of liver fat on health outcomes independent of BMI, alcohol consumption and lipids using data from published genome-wide association studies and FinnGen. RESULTS We identified 13 genetic variants associated with liver fat that had differing effects on the risks of health outcomes. Genetic variants associated with impaired hepatic triglyceride export showed liver fat-increasing alleles to be correlated with a reduced risk of coronary artery disease and myocardial infarction but an elevated risk of type 2 diabetes, while variants associated with enhanced de novo lipogenesis showed liver fat-increasing alleles to be linked to a higher risk of myocardial infarction and coronary artery disease. Genetically higher liver fat content increased the risk of non-alcohol-related cirrhosis, hepatocellular carcinoma, and intrahepatic bile duct and gallbladder cancers, exhibiting a dose-dependent relationship, irrespective of the mechanism. CONCLUSION This study provides fresh insight into the heterogeneous effect of liver fat on health outcomes. It challenges the notion that liver fat per se is an independent risk factor for cardiovascular disease, underscoring the dependency of this association on the specific mechanisms that drive fat accumulation in the liver. However, excess liver fat, regardless of the underlying mechanism, appears to be causally linked to cirrhosis and cancers in a dose-dependent manner. IMPACT AND IMPLICATION This research advances our understanding of the heterogeneity in mechanisms influencing liver fat accumulation, providing new insights into how liver fat accumulation may impact various health outcomes. The findings challenge the notion that liver fat is an independent risk factor for cardiovascular disease and highlight the mechanistic effect of some genetic variants on fat accumulation and the development of cardiovascular diseases. This study is of particular importance for healthcare professionals including physicians and researchers, as well as patients, as it allows for more targeted and personalised treatment by understanding the relationship between liver fat and various health outcomes. The findings emphasise the need for a personalised management approach and a reshaping of risk assessment criteria. It also provides room for prioritising a clinical intervention aimed at reducing liver fat content (likely via intentional weight loss) that could help protect against liver-related fibrosis and cancer.
Collapse
Affiliation(s)
- Altayeb Ahmed
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, UK
| | | | - Jimmy D Bell
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Hanieh Yaghootkar
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, UK.
| |
Collapse
|
26
|
Marek GW, Malhi H. MetALD: Does it require a different therapeutic option? Hepatology 2024; 80:1424-1440. [PMID: 38820071 DOI: 10.1097/hep.0000000000000935] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/03/2024] [Indexed: 06/02/2024]
Abstract
New guidelines for the definitions of steatotic liver disease have named the entity of metabolic dysfunction and alcohol-associated liver disease (MetALD) as an overlap condition of metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-associated liver disease. There is a broad range of therapeutics in all stages of development for MASLD, but these therapeutics, in general, have not been studied in patients with significant ongoing alcohol use. In this review, we discuss the current understanding of the endogenous and exogenous risks for MASLD and MetALD. Rational strategies for therapeutic intervention in MetALD include biopsychosocial interventions, alcohol use cessation strategies, including the use of medications for alcohol use disorder, and judicious use of therapeutics for steatotic liver disease. Therapeutics with promise for MetALD include incretin-based therapies, FGF21 agonists, thyroid hormone receptor beta agonists, sodium-glucose co-transporter 2 inhibitors, and agents to modify de novo lipogenesis. Currently, glucagon-like peptide 1 receptor agonists and peroxisome proliferator-activated receptor γ agonists have the largest body of literature supporting their use in MASLD, and there is a paucity of agents in trials for alcohol-associated liver disease. From existing studies, it is not clear if unique therapeutics or a combinatorial approach are needed for MetALD. Further elucidation of the safety and benefits of MASLD-related therapies is of paramount importance for advancing therapeutics for MetALD in carefully designed inclusive clinical trials.
Collapse
Affiliation(s)
- George W Marek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
27
|
Mak LY, Liu K, Chirapongsathorn S, Yew KC, Tamaki N, Rajaram RB, Panlilio MT, Lui R, Lee HW, Lai JCT, Kulkarni AV, Premkumar M, Lesmana CRA, Hsu YC, Huang DQ. Liver diseases and hepatocellular carcinoma in the Asia-Pacific region: burden, trends, challenges and future directions. Nat Rev Gastroenterol Hepatol 2024; 21:834-851. [PMID: 39147893 DOI: 10.1038/s41575-024-00967-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/17/2024]
Abstract
Globally, nearly half of deaths from cirrhosis and chronic liver diseases (CLD) and three-quarters of deaths from hepatocellular carcinoma (HCC) occur in the Asia-Pacific region. Chronic hepatitis B is responsible for the vast majority of liver-related deaths in the region. Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common form of CLD, affecting an estimated 30% of the adult population. Compared with people of European descent, people from the Asia-Pacific region carry more genetic variants associated with MASLD and its progression. Alcohol is a fast-growing cause of CLD and HCC in Asia as a result of the rising per-capita consumption of alcohol. Drug-induced liver injury is under-recognized and probably has a high prevalence in this region. The epidemiological and outcome data of acute-on-chronic liver failure are heterogeneous, and non-unified definitions across regions contribute to this heterogeneity. CLDs are severely underdiagnosed, and effective treatments and vaccinations are underutilized. In this Review, we highlight trends in the burden of CLD and HCC in the Asia-Pacific region and discuss the rapidly changing aetiologies of liver disease. We examine the multiple gaps in the care cascade and propose mitigating strategies and future directions.
Collapse
Affiliation(s)
- Lung-Yi Mak
- The University of Hong Kong, Hong Kong, China
| | - Ken Liu
- The University of Sydney, Sydney, Australia
| | | | | | | | | | | | - Rashid Lui
- The Chinese University of Hong Kong, Hong Kong, China
| | - Hye Won Lee
- Yonsei University College of Medicine, Seoul, Korea
| | | | - Anand V Kulkarni
- Department of Hepatology, Asian Institute of Gastroenterology, Hyderabad, India
| | - Madhumita Premkumar
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Yao Chun Hsu
- Department of Medical Research, E-Da Hospital, Kaohsiung, Taiwan; School of Medicine and Graduate Institute of Medicine, I-Shou University, Kaohsiung, Taiwan
- School of Medicine and Graduate Institute of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Daniel Q Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Division of Gastroenterology and Hepatology, National University Hospital, Singapore, Singapore.
| |
Collapse
|
28
|
Xu P, Liu M, Liu M, Shen A. Management of non-alcoholic fatty liver disease-associated hepatocellular carcinoma. Biosci Trends 2024; 18:431-443. [PMID: 39428499 DOI: 10.5582/bst.2024.01295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
In recent years, with the decline in HBV and HCV infections, there has been a corresponding reduction in both the morbidity and mortality of virus-associated HCC. Nevertheless, rising living standards, coupled with the increasing prevalence of metabolic disorders like diabetes and obesity, have led to a rapid surge in non-alcoholic fatty liver disease-associated hepatocellular carcinoma (NAFLD-HCC) incidence. The mechanisms underlying the progression from NAFLD to NAFLD-HCC are multifaceted and remain incompletely understood. Current research suggests that genetic predisposition, metabolic dysregulation, lipotoxicity, oxidative stress, and inflammation are key contributing factors. Given the complexity of these mechanisms and the frequent occurrence of metabolic comorbidities like type 2 diabetes mellitus (T2DM) and cardiovascular disease in NAFLD-HCC patients, there is a pressing need for tailored therapeutic strategies, along with novel prevention, monitoring, and treatment approaches that are personalized to the patient's pathophysiology. Due to the limited depth of research, incomplete understanding of pathogenesis, and insufficient clinical data on NAFLD-HCC treatment, current therapeutic approaches largely rely on tumor staging. In this review, we synthesize current research on the pathogenesis, surveillance, diagnosis, treatment, and prevention of NAFLD-HCC, and offer perspectives for future studies, particularly regarding its underlying mechanisms.
Collapse
Affiliation(s)
- Peijun Xu
- Department of Hepatobiliary Pancreatic Cancer Center, Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Maoyun Liu
- Department of Hepatobiliary Pancreatic Cancer Center, Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Miao Liu
- Department of Gastrointestinal Cancer Center, Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Ai Shen
- Department of Hepatobiliary Pancreatic Cancer Center, Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
29
|
Moyana TN. Metabolic dysfunction-associated steatotic liver disease: The question of long-term high-normal alanine aminotransferase as a screening test. World J Gastroenterol 2024; 30:4576-4582. [PMID: 39563746 PMCID: PMC11572615 DOI: 10.3748/wjg.v30.i42.4576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/26/2024] [Accepted: 10/09/2024] [Indexed: 10/31/2024] Open
Abstract
The growing prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is being driven by the obesity epidemic. The quest for solutions continues particularly with regard to early detection. This editorial comments on the utility of long-term high-normal alanine aminotransferase (ALT) in screening for MASLD. Chen et al found that new onset MASLD can be detected by repetitively high normal ALT. Implicit in this concept is the question of what should be the accepted upper limit of normal (ULN) for ALT. It was previously set at 40 IU/L based on studies that included people with subclinical liver disease but the new consensus is 30/19 U/L in healthy males/females. Thus, when Chen et al defines the ULN as 40 U/L, others may view it as excessively high. It is important to recognize the variables affecting ULN e.g. instrumentation, diurnal variations, exercise and ageing. These variables matter when the distinctions are subtle e.g. normal vs high-normal. In this regard, the utility of long-term high normal ALT as a disease marker could be enhanced by combining it with other biomarkers, imaging and MASLD genetics to create machine learning classifiers. All in all, Chen et al's work on long-term high normal ALT as a marker of new-onset MASLD deserves merit.
Collapse
Affiliation(s)
- Terence N Moyana
- Department of Pathology and Laboratory Medicine, University of Ottawa and The Ottawa Hospital, Ottawa K1H 8L6, Ontario, Canada
| |
Collapse
|
30
|
Kaylan KB, Paul S. NAFLD No More: A Review of Current Guidelines in the Diagnosis and Evaluation of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Curr Diab Rep 2024; 25:5. [PMID: 39535566 DOI: 10.1007/s11892-024-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW Provide a concise update on metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), as well as a practical approach to screening and initial evaluation. RECENT FINDINGS Nomenclature changes have placed a greater focus on cardiometabolic risk factors in the definition of MASLD. Screening for MASLD is by stepwise noninvasive serum and imaging tests which can identify patients at risk for advanced fibrosis and liver-related complications. MASLD has been increasing in prevalence and disease burden but is underrecognized in primary care and endocrinology clinics. Multiple society guidelines, synthesized here, provide a framework for the initial approach in the diagnosis and evaluation of MASLD. Recent advances in pharmacologic treatment underline the importance of screening for patients who are at risk for advanced fibrosis as they are most likely to benefit from new drug classes, such as the liver-directed thyroid receptor agonist resmiterom.
Collapse
Affiliation(s)
- Kerim B Kaylan
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Medicine, Chicago, IL, USA
| | - Sonali Paul
- Section of Gastroenterology, Hepatology, and Nutrition, Center for Liver Diseases, The University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
31
|
Küçük K, Moreno C, Nijmi H, Daoud M, Mintoff D, Willaert F, Benhadou F. Identification of Risk Factors Associated with Metabolic Dysfunction-Associated Steatotic Liver Disease in Psoriatic Patients. Dermatology 2024; 241:92-100. [PMID: 39536736 DOI: 10.1159/000541796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 09/28/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) is a common cause of chronic liver disease. Patients suffering from psoriasis are at an increased risk of developing MASLD. Psoriasis and MASLD share a pro-inflammatory cytokine milieu; however, it is still unclear whether these conditions are related through shared metainflammatory processes or shared comorbidities such as obesity, diabetes, insulin resistance, and metabolic syndrome. The aim of our study was to better characterize the anthropometric and metabolic profile of psoriatic patients with MASLD. METHODS We conducted a prospective, single-center, cross-sectional study between June 2014 and August 2017. Recruitment was restricted to adult patients with psoriasis. Blood analysis, liver ultrasonography, and a FibroScan were performed. Blood investigations, baseline anthropometric measurements, and components of fatty liver disease (hepatic ultrasound, FibroScan) were assessed. RESULTS A total of 100 patients were recruited, of which, 43% (65.1% men, n = 28) were diagnosed with MASLD. The mean BMI was significantly higher in MASLD than in non-MASLD (27.7 kg/m2 vs. 30.1 kg/m2, p =< 0.001). The mean waist circumference in MASLD patients was significantly higher than in non-MASLD patients (105.6 cm vs. 97.2 cm, p = 0.005). There was no significant difference between the mean age of both patient groups (50.4 vs. 47.3 years, p = 0.26). Psoriatic arthritis was more prevalent in MASLD than in the non-MASLD group (14.3% vs. 1.8%, p = 0.004). Biochemical analysis revealed significantly higher C-peptide level in patients with MASLD compared with patients without MASLD (2.5 vs. 1.6 ng/mL, p = 0.036). Moreover, MASLD patients were found to have a lower HDL level and higher glycemia, triglyceridemia, cholesterol, and LDL levels than non-MASLD patients. A total of 16.3% of patients with MASLD had fibrosis stage ranging from F2 to F4 based on liver stiffness measurement compared with only 10.6% of patients without MASLD. DISCUSSION We identified parameters which were more prevalent in patients with psoriasis having MASLD, specifically a high BMI, elevated triglyceride levels, decreased HDL levels, and an elevated level of C-peptide. Patients with psoriasis and MASLD were more likely to suffer from comorbid psoriatic arthritis, despite having similar psoriasis disease severity as measured by PASI. CONCLUSION This study highlights the importance of screening patients with psoriasis for MASLD to prevent the progression to liver fibrosis.
Collapse
Affiliation(s)
- Kirley Küçük
- Department of Dermatology, Hôpitaux Universitaires de Bruxelles (HUB), Université libre de Bruxelles, Brussels, Belgium
| | - Christophe Moreno
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, CUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Hassane Nijmi
- Department of Dermatology, Hôpitaux Universitaires de Bruxelles (HUB), Université libre de Bruxelles, Brussels, Belgium
| | - Mathieu Daoud
- Department of Dermatology, Hôpitaux Universitaires de Bruxelles (HUB), Université libre de Bruxelles, Brussels, Belgium
| | - Dillon Mintoff
- Department of Dermatology, Mater Dei Hospital, Msida, Malta
| | - Fabienne Willaert
- Department of Dermatology, Hôpitaux Universitaires de Bruxelles (HUB), Université libre de Bruxelles, Brussels, Belgium
| | - Farida Benhadou
- Department of Dermatology, Hôpitaux Universitaires de Bruxelles (HUB), Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
32
|
Israelsen M, Francque S, Tsochatzis EA, Krag A. Steatotic liver disease. Lancet 2024; 404:1761-1778. [PMID: 39488409 DOI: 10.1016/s0140-6736(24)01811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 11/04/2024]
Abstract
Steatotic liver disease is the overarching term for conditions characterised by abnormal lipid accumulation in the liver (liver or hepatic steatosis). Steatotic liver disease encompasses what was previously termed non-alcoholic fatty liver disease (NAFLD), which is now called metabolic dysfunction-associated steatotic liver disease (MASLD). Additionally, steatotic liver disease includes alcohol-related liver disease (ALD) and MetALD, the new classification for the overlap between MASLD and ALD, and rare causes of liver steatosis. Cirrhosis is globally the 11th leading cause of death, and steatotic liver disease has become the leading cause of cirrhosis in the EU and USA. Steatotic liver disease affects around 30% of the global population and is mainly driven by obesity, type 2 diabetes, and alcohol intake, but only a minor proportion with steatotic liver disease progress to cirrhosis. The presence and progression of liver fibrosis led by hepatic inflammation is the main predictor of liver-related death across the entire spectrum of steatotic liver diseases. A combination of recent advancements of widely available biomarkers for early detection of liver fibrosis together with considerable advancements in therapeutic interventions offer the possibility to reduce morbidity and mortality in patients with steatotic liver disease. This Seminar covers the recent reclassification of steatotic liver disease and how it reflects clinical practice and prognosis. For early detection of liver fibrosis, we propose a collaborative diagnostic framework between primary care and liver specialists. Lastly, we discuss current best practices for managing steatotic liver disease, we explore therapeutic targets across the spectrum of steatotic liver diseases, and we review the pipeline of drugs in development for MASLD.
Collapse
Affiliation(s)
- Mads Israelsen
- Centre for Liver Research and Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; InflaMed Centre of Excellence, Translational Sciences in Inflammation and Immunology, University of Antwerp, Antwerp, Belgium
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College of London, London, UK
| | - Aleksander Krag
- Centre for Liver Research and Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Institute of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
33
|
Pelusi S, Ronzoni L, Rondena J, Rosso C, Pennisi G, Dongiovanni P, Margarita S, Carpani R, Soardo G, Prati D, Cespiati A, Petta S, Bugianesi E, Valenti L. Prevalence and Determinants of Liver Disease in Relatives of Italian Patients With Advanced MASLD. Clin Gastroenterol Hepatol 2024; 22:2231-2239.e4. [PMID: 38216023 DOI: 10.1016/j.cgh.2023.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction associated steatotic liver disease (MASLD) has a strong genetic component. The aim of this study was to examine noninvasively the prevalence of MASLD and of advanced fibrosis in relatives of patients with advanced MASLD and the risk factors for liver involvement, with a focus on the contribution of common genetic risk variants. METHODS We prospectively enrolled 98 consecutive probands with advanced fibrosis and/or hepatocellular carcinoma caused by MASLD and 160 nontwin first-degree relatives noninvasively screened for MASLD and advanced fibrosis at 4 Italian centers. We evaluated common genetic determinants and polygenic risk scores of liver disease. RESULTS Among relatives, prevalence of MASLD was 56.8% overall, whereas advanced fibrosis was observed in 14.4%. At multivariable analysis in relatives, MASLD was associated with body mass index (odds ratio [OR], 1.31 [1.18-1.46]) and tended to be associated with diabetes (OR, 5.21 [0.97-28.10]), alcohol intake (OR, 1.32 [0.98-1.78]), and with female sex (OR, 0.54 [0.23-1.15]), whereas advanced fibrosis was associated with diabetes (OR, 3.13 [1.16-8.45]) and nearly with body mass index (OR, 1.09 [1.00-1.19]). Despite that the PNPLA3 risk variant was enriched in probands (P = .003) and overtransmitted to relatives with MASLD (P = .045), evaluation of genetic risk variants and polygenic risk scores was not useful to guide noninvasive screening of advanced fibrosis in relatives. CONCLUSIONS We confirmed that about 1 in 7 relatives of patients with advanced MASLD has advanced fibrosis, supporting clinical recommendations to perform family screening in this setting. Genetic risk variants contributed to liver disease within families but did not meaningfully improve fibrosis risk stratification.
Collapse
Affiliation(s)
- Serena Pelusi
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Ronzoni
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jessica Rondena
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Rosso
- Division of Gastroenterology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Grazia Pennisi
- Department of Health promotion, Mother and Childcare, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Margarita
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Rossana Carpani
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Soardo
- Clinica Medica-Liver Unit, Department of Medicine, University of Udine, Udine, Italy; Italian Liver Foundation, Area Science Park, Basovizza-Trieste, Italy
| | - Daniele Prati
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Annalisa Cespiati
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Salvatore Petta
- Department of Health promotion, Mother and Childcare, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Elisabetta Bugianesi
- Division of Gastroenterology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
34
|
Moonlisarn K, Somnark P, Boonkaew B, Bunchorntavakul C, Tangkijvanich P. Interaction Between PNPLA3 and SIRT5 Genetic Variants in Association with Liver Fibrosis Severity in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease. Genes (Basel) 2024; 15:1370. [PMID: 39596570 PMCID: PMC11593416 DOI: 10.3390/genes15111370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES This study evaluated the association between polymorphisms in the PNPLA3, TM6SF2, HSD17B13, and SIRT5 genes and the severity of fibrosis and steatosis in metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Fibrosis and steatosis were assessed by MRE and MRI-PDFF, respectively. The polymorphisms were determined by allelic discrimination in blood samples. RESULTS 204 patients aged 57.0 ± 13.5 years were included. Sixty-two (30.4%) patients had significant fibrosis (≥F2). Among F2-F4 fibrosis, the PNPLA3 rs738409 GG genotype was significantly higher than the CC + CG genotypes (44.9% vs. 21.4%, p = 0.001). The SIRT5 rs12216101 GG vs. TT + TG genotypes also exhibited a similar trend (64.3% vs. 27.9%, p = 0.012). In multivariate analysis, the PNPLA3 GG genotype (OR = 3.48, 95%CI: 1.50-8.06; p = 0.004) and SIRT5 rs12216101 GG genotype (OR = 5.43, 95%CI: 1.32-22.33; p = 0.019) were independently associated with F2-F4 fibrosis. Additionally, the proportion of patients with F2-F4 fibrosis significantly increased with the number of combined risk genotypes. Among S2-S3 steatosis, the prevalence of HSD17B13 AG + GG genotypes was higher than that of the AA genotype (37.5% vs. 23.9%, p = 0.048) and independently associated with moderate/severe steatosis in multivariate analysis (OR = 2.26, 95%CI: 1.14-4.49; p = 0.020). CONCLUSIONS Our data indicate that the PNPLA3 and SIRT5 polymorphisms were independently and additively linked to significant fibrosis, while the HSD17B13 polymorphism was associated with increased steatosis in Thai populations. These data might emphasize the importance of genetic variants in progressive MASLD.
Collapse
Affiliation(s)
- Kamonchanok Moonlisarn
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (P.S.); (B.B.)
| | - Pornjira Somnark
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (P.S.); (B.B.)
| | - Bootsakorn Boonkaew
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (P.S.); (B.B.)
| | | | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (K.M.); (P.S.); (B.B.)
| |
Collapse
|
35
|
Lee DW, Oh J, Lee YM, Bae HJ, Lim YH. Association between heavy metal exposure and biomarkers for non-alcoholic fatty liver disease in Korean adolescents. Heliyon 2024; 10:e37840. [PMID: 39386834 PMCID: PMC11462472 DOI: 10.1016/j.heliyon.2024.e37840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Objectives The global prevalence of non-alcoholic fatty liver disease (NAFLD) in adolescents has increased. In addition to childhood obesity, environmental risk factors, such as heavy metals that are known to be involved in hepatotoxicity, play role in NAFLD occurrence. However, their association with NAFLD remains unclear. This study aimed to investigate the association between heavy metal exposure and NAFLD biomarkers in adolescents. Methods In this cross-sectional study, we used the data of a total of 1505 adolescents aged 12-17 years who participated in the Korean National Environmental Health Survey III (2015-2017) and IV (2018-2020). The presence of blood lead (BPb), blood mercury (BHg), urinary mercury (UHg), and urinary cadmium (UCd) were measured. Liver enzymes including serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and gamma-glutamyl transferase (GGT) were evaluated. For NAFLD biomarkers, the hepatic steatosis index (HSI) was calculated. Multivariate linear regression models, weighted quantile sum (WQS) regression, and Bayesian kernel machine regression (BKMR) model were used to investigate the association between heavy metals and NAFLD biomarkers. Results Among heavy metals, mercury presence showed a significant association with NAFLD biomarkers. Two-fold increases in BHg and UHg were associated with 0.21 points (95 % confidence interval [CI]: 0.08-0.35) and 0.19 points (95 % CI: 0.09-0.30) higher HSI, respectively. In the WQS model, heavy metal mixture was significantly associated with increased HSI (β = 0.06, 95 % CI: 0.01-0.11). Similarly, in the BKMR model, heavy metal mixture was positively associated with NAFLD biomarkers, and BHg was the most important contributor in the association. Conclusions BHg and UHg were significantly associated with NAFLD biomarkers in adolescents, indicating that organic and inorganic mercury exposure could potentially be a risk factor for NAFLD. To mitigate and address the risk of NAFLD associated with heavy metal exposure, it is imperative to take measure to reduce avoidable mercury exposure is necessary.
Collapse
Affiliation(s)
- Dong-Wook Lee
- Department of Occupational and Environmental Medicine, Inha University Hospital, Inha University, Incheon, Republic of Korea
| | - Jongmin Oh
- Institute of Ewha-SCL for Environmental Health (IESEH), Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Yu Min Lee
- Department of Occupational and Environmental Medicine, Severance Hospital, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Hyun-Joo Bae
- Korea Environment Institute, Sejong, Republic of Korea
| | - Youn-Hee Lim
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Moretti V, Romeo S, Valenti L. The contribution of genetics and epigenetics to MAFLD susceptibility. Hepatol Int 2024; 18:848-860. [PMID: 38662298 PMCID: PMC11450136 DOI: 10.1007/s12072-024-10667-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/25/2024] [Indexed: 04/26/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common liver disease worldwide. The risk of developing MAFLD varies among individuals, due to a combination of environmental inherited and acquired genetic factors. Genome-wide association and next-generation sequencing studies are leading to the discovery of the common and rare genetic determinants of MAFLD. Thanks to the great advances in genomic technologies and bioinformatics analysis, genetic and epigenetic factors involved in the disease can be used to develop genetic risk scores specific for liver-related complications, which can improve risk stratification. Genetic and epigenetic factors lead to the identification of specific sub-phenotypes of MAFLD, and predict the individual response to a pharmacological therapy. Moreover, the variant transcripts and protein themselves represent new therapeutic targets. This review will discuss the current status of research into genetic as well as epigenetic modifiers of MAFLD development and progression.
Collapse
Affiliation(s)
- Vittoria Moretti
- Precision Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Via F Sforza 35, 20122, Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Via F Sforza 35, 20122, Milan, Italy.
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
37
|
Marchetti A, Pelusi S, Marella A, Malvestiti F, Ricchiuti A, Ronzoni L, Lionetti M, Moretti V, Bugianesi E, Miele L, Vespasiani-Gentilucci U, Dongiovanni P, Federico A, Soardo G, D’Ambrosio R, McCain MV, Reeves HL, La Mura V, Prati D, Bolli N, Valenti L. Impact of clonal hematopoiesis of indeterminate potential on hepatocellular carcinoma in individuals with steatotic liver disease. Hepatology 2024; 80:816-827. [PMID: 38470216 PMCID: PMC11407776 DOI: 10.1097/hep.0000000000000839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a global epidemic and is the most rapidly rising cause of HCC. Clonal hematopoiesis of indeterminate potential (CHIP) contributes to neoplastic and cardiometabolic disorders and is considered a harbinger of tissue inflammation. CHIP was recently associated with increased risk of liver disease. The aim of this study was to examine whether CHIP is associated with HCC development in patients with SLD. APPROACH AND RESULTS We considered individuals with MASLD-HCC (n=208) and controls with (n =414) and without (n =259) advanced fibrosis who underwent whole exome sequencing. CHIP was diagnosed when ≥2 variant callers identified a known myeloid mutation with variant allele frequency ≥2%. CHIP was observed in 116 participants (13.1%), most frequently in DNMT3A, TET2, TP53 , and ASXL1 , and correlated with age ( p <0.0001) and advanced liver fibrosis (p=0.001). Higher aspartate aminotransferase levels predicted non- DNMT3A -CHIP, in particular with variant allele frequency ≥10% (OR: 1.14, 1.03 -1.28 and OR: 1.30, 1.12 -1.49, respectively, p <0.05). After adjustment for sex, diabetes, and a polygenic risk, a score of inherited MASLD predisposition CHIP was associated with cirrhosis (2.00, 1.30 -3.15, p =0.02), and with HCC even after further adjustment for cirrhosis (OR: 1.81, 1.11 -2.00, 1.30 -3.15, p =0.002). Despite the strong collinearity among aging and development of CHIP and HCC, non- DNTM3A -CHIP, and TET2 lesions remained associated with HCC after full correction for clinical/genetics covariates and age (OR: 2.45, 1.35 -4.53; OR: 4.8, 1.60 -17.0, p =0.02). CONCLUSIONS We observed an independent association between CHIP, particularly related to non- DNTM3A and TET2 genetic lesions and MASLD-HCC.
Collapse
Affiliation(s)
- Alfredo Marchetti
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Serena Pelusi
- Transfusion Medicine Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessio Marella
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Francesco Malvestiti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Antony Ricchiuti
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Ronzoni
- Transfusion Medicine Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Lionetti
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Vittoria Moretti
- Transfusion Medicine Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Turin, Italy
| | - Luca Miele
- Dipartimento Universitario Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Area Medicina Interna, Gastroenterologia e Oncologia Medica, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Clinical Medicine and Hepatology Unit, Department of Medicine and Surgery, Campus Bio-Medico University of Rome, Rome, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandro Federico
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giorgio Soardo
- Department of Medicine, Clinica Medica, European Excellence Center for Arterial Hypertension, University of Udine, Udine, Italy
| | - Roberta D’Ambrosio
- Gastroenterology and Hepatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Misti V. McCain
- Newcastle University Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen L. Reeves
- Newcastle University Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Vincenzo La Mura
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- General Medicine, Haemostasis and Thrombosis, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniele Prati
- Transfusion Medicine Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Transfusion Medicine Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Biological Resource Center Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
38
|
Allen AM, Younossi ZM, Diehl AM, Charlton MR, Lazarus JV. Envisioning how to advance the MASH field. Nat Rev Gastroenterol Hepatol 2024; 21:726-738. [PMID: 38834817 DOI: 10.1038/s41575-024-00938-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
Since 1980, the cumulative effort of scientists and health-care stakeholders has advanced the prerequisites to address metabolic dysfunction-associated steatotic liver disease (MASLD), a prevalent chronic non-communicable liver disease. This effort has led to, among others, the approval of the first drug specific for metabolic dysfunction-associated steatohepatitis (MASH; formerly known as nonalcoholic steatohepatitis). Despite substantial progress, MASLD is still a leading cause of advanced chronic liver disease, including primary liver cancer. This Perspective contextualizes the nomenclature change from nonalcoholic fatty liver disease to MASLD and proposes important considerations to accelerate further progress in the field, optimize patient-centric multidisciplinary care pathways, advance pharmacological, behavioural and diagnostic research, and address health disparities. Key regulatory and other steps necessary to optimize the approval and access to upcoming additional pharmacological therapeutic agents for MASH are also outlined. We conclude by calling for increased education and awareness, enhanced health system preparedness, and concerted action by policy-makers to further the public health and policy agenda to achieve at least parity with other non-communicable diseases and to aid in growing the community of practice to reduce the human and economic burden and end the public health threat of MASLD and MASH by 2030.
Collapse
Affiliation(s)
- Alina M Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zobair M Younossi
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, VA, USA
- The Global NASH Council, Washington DC, USA
| | | | - Michael R Charlton
- Center for Liver Diseases, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jeffrey V Lazarus
- The Global NASH Council, Washington DC, USA.
- CUNY Graduate School of Public Health and Health Policy (CUNY SPH), New York, NY, USA.
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain.
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
39
|
Tang P, Keshi E, Wilken S, Wutsdorff L, Mougnekabol J, Pratschke J, Sauer IM, Haep N. Generation of an induced pluripotent stem cell (iPSC) line (EXSURGi001-A) from a patient homozygous for the p.Ala165Thr mutation in the MTARC1 gene. Stem Cell Res 2024; 80:103516. [PMID: 39094505 DOI: 10.1016/j.scr.2024.103516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/23/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD), the leading cause of end-stage liver disease in developed countries, is expected to increase over the next decade. Characterized by hepatic steatosis, MAFLD is commonly studied in animal models. Here, we generated a human induced pluripotent stem cell (iPSC) line from a patient homozygous of the protective MTARC1 gene variant rs2642438:A. This line displays a normal karyotype and typical pluripotent stem cell morphology and can differentiate into all three germ layers in vitro.
Collapse
Affiliation(s)
- Peter Tang
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Eriselda Keshi
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Clinician Scientist Program, Berlin Institute of Health at Charité (BIH), Anna-Louisa-Karsch-Str. 2, 10178 Berlin, Germany
| | - Silvana Wilken
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Louise Wutsdorff
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Julienne Mougnekabol
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Igor M Sauer
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nils Haep
- Department of Surgery, Campus Virchow-Klinikum, Experimental Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Clinician Scientist Program, Berlin Institute of Health at Charité (BIH), Anna-Louisa-Karsch-Str. 2, 10178 Berlin, Germany.
| |
Collapse
|
40
|
Martín Barraza JI, Bars-Cortina D. Dietary Pattern's Role in Hepatic Epigenetic and Dietary Recommendations for the Prevention of NAFLD. Nutrients 2024; 16:2956. [PMID: 39275272 PMCID: PMC11396970 DOI: 10.3390/nu16172956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
NAFLD has emerged as a significant public health concern, with its prevalence increasing globally. Emphasizing the complex relationship between dietary patterns and epigenetic modifications such as DNA methylation or miRNA expression can exert a positive impact on preventing and managing metabolic disorders, including NAFLD, within the 2030 Sustainable Development Goals. This review aims to evaluate the influence of dietary patterns on hepatic epigenetic gene modulation and provide dietary recommendations for the prevention and management of NAFLD in the general population. METHODS Comprehensive screening and eligibility criteria identified eleven articles focusing on epigenetic changes in NAFLD patients through dietary modifications or nutrient supplementation. RESULTS AND DISCUSSION Data were organized based on study types, categorizing them into evaluations of epigenetic changes in NAFLD patients through dietary pattern modifications or specific nutrient intake. CONCLUSIONS The study highlights the importance of dietary interventions in managing and preventing NAFLD, emphasizing the potential of dietary patterns to influence hepatic epigenetic gene modulation. This study provides valuable insights and recommendations to mitigate the risk of developing NAFLD: (i) eat a primarily plant-based diet; (ii) increase consumption of high-fiber foods; (iii) consume more polyunsaturated and monounsaturated fatty acids; (iv) limit processed foods, soft drinks, added sugars, and salt; and (v) avoid alcohol.
Collapse
Affiliation(s)
| | - David Bars-Cortina
- Oncology Data Analytics Program (ODAP), Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), L’Hospitalet del Llobregat, 08908 Barcelona, Spain
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
- Department of Health Sciences, Universitat Oberta de Catalunya, 08018 Barcelona, Spain
| |
Collapse
|
41
|
Tacke F, Horn P, Wai-Sun Wong V, Ratziu V, Bugianesi E, Francque S, Zelber-Sagi S, Valenti L, Roden M, Schick F, Yki-Järvinen H, Gastaldelli A, Vettor R, Frühbeck G, Dicker D. EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol 2024; 81:492-542. [PMID: 38851997 DOI: 10.1016/j.jhep.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 06/10/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
42
|
Faraggi E, Jernigan RL, Kloczkowski A. Rapid discrimination between deleterious and benign missense mutations in the CAGI 6 experiment. Hum Genomics 2024; 18:89. [PMID: 39192324 PMCID: PMC11350969 DOI: 10.1186/s40246-024-00655-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/08/2024] [Indexed: 08/29/2024] Open
Abstract
We describe the machine learning tool that we applied in the CAGI 6 experiment to predict whether single residue mutations in proteins are deleterious or benign. This tool was trained using only single sequences, i.e., without multiple sequence alignments or structural information. Instead, we used global characterizations of the protein sequence. Training and testing data for human gene mutations was obtained from ClinVar (ncbi.nlm.nih.gov/pub/ClinVar/), and for non-human gene mutations from Uniprot (www.uniprot.org). Testing was done on post-training data from ClinVar. This testing yielded high AUC and Matthews correlation coefficient (MCC) for well trained examples but low generalizability. For genes with either sparse or unbalanced training data, the prediction accuracy is poor. The resulting prediction server is available online at http://www.mamiris.com/Shoni.cagi6.
Collapse
Affiliation(s)
- Eshel Faraggi
- Research and Information Systems, LLC, 1620 E. 72nd ST., Indianapolis, IN, 46240, USA.
- Physics Department, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| | - Robert L Jernigan
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, 50011, USA
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Columbus, OH, 43205, USA
- Battelle Center for Mathematical Medicine, The Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, 43205, USA
| |
Collapse
|
43
|
Habib S. Team players in the pathogenesis of metabolic dysfunctions-associated steatotic liver disease: The basis of development of pharmacotherapy. World J Gastrointest Pathophysiol 2024; 15:93606. [PMID: 39220834 PMCID: PMC11362842 DOI: 10.4291/wjgp.v15.i4.93606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Nutrient metabolism is regulated by several factors. Social determinants of health with or without genetics are the primary regulator of metabolism, and an unhealthy lifestyle affects all modulators and mediators, leading to the adaptation and finally to the exhaustion of cellular functions. Hepatic steatosis is defined by presence of fat in more than 5% of hepatocytes. In hepatocytes, fat is stored as triglycerides in lipid droplet. Hepatic steatosis results from a combination of multiple intracellular processes. In a healthy individual nutrient metabolism is regulated at several steps. It ranges from the selection of nutrients in a grocery store to the last step of consumption of ATP as an energy or as a building block of a cell as structural component. Several hormones, peptides, and genes have been described that participate in nutrient metabolism. Several enzymes participate in each nutrient metabolism as described above from ingestion to generation of ATP. As of now several publications have revealed very intricate regulation of nutrient metabolism, where most of the regulatory factors are tied to each other bidirectionally, making it difficult to comprehend chronological sequence of events. Insulin hormone is the primary regulator of all nutrients' metabolism both in prandial and fasting states. Insulin exerts its effects directly and indirectly on enzymes involved in the three main cellular function processes; metabolic, inflammation and repair, and cell growth and regeneration. Final regulators that control the enzymatic functions through stimulation or suppression of a cell are nuclear receptors in especially farnesoid X receptor and peroxisome proliferator-activated receptor/RXR ligands, adiponectin, leptin, and adiponutrin. Insulin hormone has direct effect on these final modulators. Whereas blood glucose level, serum lipids, incretin hormones, bile acids in conjunction with microbiota are intermediary modulators which are controlled by lifestyle. The purpose of this review is to overview the key players in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) that help us understand the disease natural course, risk stratification, role of lifestyle and pharmacotherapy in each individual patient with MASLD to achieve personalized care and target the practice of precision medicine. PubMed and Google Scholar databases were used to identify publication related to metabolism of carbohydrate and fat in states of health and disease states; MASLD, cardiovascular disease and cancer. More than 1000 publications including original research and review papers were reviewed.
Collapse
Affiliation(s)
- Shahid Habib
- Department of Hepatology, Liver Institute PLLC, Tucson, AZ 85712, United States
| |
Collapse
|
44
|
Wu H, Wei J, Wang S, Chen L, Zhang J, Wang N, Tan X. Dietary pattern modifies the risk of MASLD through metabolomic signature. JHEP Rep 2024; 6:101133. [PMID: 39081700 PMCID: PMC11286987 DOI: 10.1016/j.jhepr.2024.101133] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 08/02/2024] Open
Abstract
Background & Aims The EAT-Lancet Commission in 2019 advocated a plant-centric diet for health and environmental benefits, but its relation to metabolic dysfunction-associated steatotic liver disease (MASLD) is unclear. We aimed to discover the metabolite profile linked to the EAT-Lancet diet and its association with MASLD risk, considering genetic predisposition. Methods We analyzed data from 105,752 UK Biobank participants with detailed dietary and metabolomic information. We constructed an EAT-Lancet diet index and derived a corresponding metabolomic signature through elastic net regression. A weighted polygenic risk score for MASLD was computed from associated risk variants. The Cox proportional hazards model was employed to estimate hazard ratios (HRs) and 95% CIs for the risk of MASLD (defined as hospital admission or death). Results During a median follow-up period of 11.6 years, 1,138 cases of MASLD were documented. Participants in the highest group for the EAT-Lancet diet index had a multivariable HR of 0.79 (95% CI 0.66-0.95) for MASLD compared to the lowest group. The diet's impact was unaffected by genetic predisposition to MASLD (p = 0.42). Moreover, a robust correlation was found between the metabolomic signature and the EAT-Lancet diet index (Pearson r = 0.29; p <0.0001). Participants in the highest group for the metabolomic signature had a multivariable HR of 0.46 (95% CI 0.37-0.58) for MASLD, in comparison to those in the lowest group. Conclusions Higher intake of the EAT-Lancet diet and its associated metabolite signature are both linked to a reduced risk of MASLD, independently of traditional risk factors. Impact and implications Our analysis leveraging the UK Biobank study showed higher adherence to the EAT-Lancet diet was associated with a reduced risk of metabolic dysfunction-associated steatotic liver disease (MASLD). We identified a unique metabolite signature comprising 81 metabolites associated with the EAT-Lancet diet, potentially underlying the diet's protective mechanism against MASLD. These findings suggest the EAT-Lancet diet may offer substantial protective benefits against MASLD.
Collapse
Affiliation(s)
- Hanzhang Wu
- Department of Big Data in Health Science, Zhejiang University School of Public Health, Hangzhou, China. Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Jiahe Wei
- Department of Big Data in Health Science, Zhejiang University School of Public Health, Hangzhou, China. Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Shuai Wang
- Department of Big Data in Health Science, Zhejiang University School of Public Health, Hangzhou, China. Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jihui Zhang
- Center for Sleep and Circadian Medicine, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health, Hangzhou, China. Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
45
|
Manusov EG, Diego VP, Almeida M, Ortiz D, Curran JE, Galan J, Leandro AC, Laston S, Blangero J, Williams-Blangero S. Genotype-by-Environment Interactions in Nonalcoholic Fatty Liver Disease and Chronic Illness among Mexican Americans: The Role of Acculturation Stress. Genes (Basel) 2024; 15:1006. [PMID: 39202366 PMCID: PMC11353877 DOI: 10.3390/genes15081006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
This study examines the complex interplay of genetic and environmental interactions that shape chronic illness risk. Evidence is mounting for the role of genetic expression and the immune response in the pathogenesis of chronic disease. In the Rio Grande Valley of south Texas, where 90% of the population is Mexican American, chronic illnesses (including obesity, diabetes, nonalcoholic liver disease, and depression) are reaching epidemic proportions. This study leverages an ongoing family study of the genetic determinants of risk for obesity, diabetes, hypertension, hyperlipidemia, and depression in a Mexican American population. Data collected included blood pressure, BMI, hepatic transaminases, HbA1c, depression (BDI-II), acculturation/marginalization (ARSMA-II), and liver health as assessed by elastography. Heritability and genotype-by-environment (G×E) interactions were analyzed, focusing on the marginalization/separation measure of the ARSMA-II. Significant heritabilities were found for traits such as HbA1c (h2 = 0.52), marginalization (h2 = 0.30), AST (h2 = 0.25), ALT (h2 = 0.41), and BMI (h2 = 0.55). Genotype-by-environment interactions were significant for HbA1c, AST/ALT ratio, BDI-II, and CAP, indicating that genetic factors interact with marginalization to influence these traits. This study found that acculturation stress exacerbates the genetic response to chronic illness. These findings underscore the importance of considering G×E interactions in understanding disease susceptibility and may inform targeted interventions for at-risk populations. Further research is warranted to elucidate the underlying molecular pathways and replicate these findings in diverse populations.
Collapse
Affiliation(s)
- Eron G. Manusov
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Vincent P. Diego
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
| | - Marcio Almeida
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - David Ortiz
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA;
| | - Joanne E. Curran
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Jacob Galan
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Ana C. Leandro
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Sandra Laston
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Sarah Williams-Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA (J.E.C.)
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| |
Collapse
|
46
|
Lee SHT, Garske KM, Arasu UT, Kar A, Miao Z, Alvarez M, Koka A, Darci-Maher N, Benhammou JN, Pan DZ, Örd T, Kaminska D, Männistö V, Heinonen S, Wabitsch M, Laakso M, Agopian VG, Pisegna JR, Pietiläinen KH, Pihlajamäki J, Kaikkonen MU, Pajukanta P. Single nucleus RNA-sequencing integrated into risk variant colocalization discovers 17 cell-type-specific abdominal obesity genes for metabolic dysfunction-associated steatotic liver disease. EBioMedicine 2024; 106:105232. [PMID: 38991381 PMCID: PMC11663762 DOI: 10.1016/j.ebiom.2024.105232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Abdominal obesity increases the risk for non-alcoholic fatty liver disease (NAFLD), now known as metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS To elucidate the directional cell-type level biological mechanisms underlying the association between abdominal obesity and MASLD, we integrated adipose and liver single nucleus RNA-sequencing and bulk cis-expression quantitative trait locus (eQTL) data with the UK Biobank genome-wide association study (GWAS) data using colocalization. Then we used colocalized cis-eQTL variants as instrumental variables in Mendelian randomization (MR) analyses, followed by functional validation experiments on the target genes of the cis-eQTL variants. FINDINGS We identified 17 colocalized abdominal obesity GWAS variants, regulating 17 adipose cell-type marker genes. Incorporating these 17 variants into MR discovers a putative tissue-of-origin, cell-type-aware causal effect of abdominal obesity on MASLD consistently with multiple MR methods without significant evidence for pleiotropy or heterogeneity. Single cell data confirm the adipocyte-enriched mean expression of the 17 genes. Our cellular experiments across human adipogenesis identify risk variant -specific epigenetic and transcriptional mechanisms. Knocking down two of the 17 genes, PPP2R5A and SH3PXD2B, shows a marked decrease in adipocyte lipidation and significantly alters adipocyte function and adipogenesis regulator genes, including DGAT2, LPL, ADIPOQ, PPARG, and SREBF1. Furthermore, the 17 genes capture a characteristic MASLD expression signature in subcutaneous adipose tissue. INTERPRETATION Overall, we discover a significant cell-type level effect of abdominal obesity on MASLD and trace its biological effect to adipogenesis. FUNDING NIH grants R01HG010505, R01DK132775, and R01HL170604; the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (Grant No. 802825), Academy of Finland (Grants Nos. 333021), the Finnish Foundation for Cardiovascular Research the Sigrid Jusélius Foundation and the Jane and Aatos Erkko Foundation; American Association for the Study of Liver Diseases (AASLD) Advanced Transplant Hepatology award and NIH/NIDDK (P30DK41301) Pilot and Feasibility award; NIH/NIEHS F32 award (F32ES034668); Finnish Diabetes Research Foundation, Kuopio University Hospital Project grant (EVO/VTR grants 2005-2021), the Academy of Finland grant (Contract no. 138006); Academy of Finland (Grant Nos 335443, 314383, 272376 and 266286), Sigrid Jusélius Foundation, Finnish Medical Foundation, Finnish Diabetes Research Foundation, Novo Nordisk Foundation (#NNF20OC0060547, NNF17OC0027232, NNF10OC1013354) and Government Research Funds to Helsinki University Hospital; Orion Research Foundation, Maud Kuistila Foundation, Finish Medical Foundation, and University of Helsinki.
Collapse
Affiliation(s)
- Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Uma Thanigai Arasu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nicholas Darci-Maher
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jihane N Benhammou
- Vatche and Tamar Manoukian Division of Digestive Diseases and Gastroenterology, Hepatology and Parenteral Nutrition, David Geffen School of Medicine at UCLA and VA Greater Los Angeles HCS, Los Angeles, CA, USA
| | - David Z Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Tiit Örd
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Ville Männistö
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Internal Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Vatche G Agopian
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joseph R Pisegna
- Department of Medicine and Human Genetics, Division of Gastroenterology, Hepatology and Parenteral Nutrition, David Geffen School of Medicine at UCLA and VA Greater Los Angeles HCS, Los Angeles, CA, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Healthy WeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Minna U Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
47
|
Thakral N, Desalegn H, Diaz LA, Cabrera D, Loomba R, Arrese M, Arab JP. A Precision Medicine Guided Approach to the Utilization of Biomarkers in MASLD. Semin Liver Dis 2024; 44:273-286. [PMID: 38991536 DOI: 10.1055/a-2364-2928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
The new nomenclature of metabolic dysfunction-associated steatotic liver disease (MASLD) emphasizes a positive diagnosis based on cardiometabolic risk factors. This definition is not only less stigmatizing but also allows for subclassification and stratification, thereby addressing the heterogeneity of what was historically referred to as nonalcoholic fatty liver disease. The heterogeneity within this spectrum is influenced by several factors which include but are not limited to demographic/dietary factors, the amount of alcohol use and drinking patterns, metabolic status, gut microbiome, genetic predisposition together with epigenetic factors. The net effect of this dynamic and intricate system-level interaction is reflected in the phenotypic presentation of MASLD. Therefore, the application of precision medicine in this scenario aims at complex phenotyping with consequent individual risk prediction, development of individualized preventive strategies, and improvements in the clinical trial designs. In this review, we aim to highlight the importance of precision medicine approaches in MASLD, including the use of novel biomarkers of disease, and its subsequent utilization in future study designs.
Collapse
Affiliation(s)
- Nimish Thakral
- Division of Gastroenterology and Hepatology, University of Kentucky, Lexington, Kentucky
| | - Hailemichael Desalegn
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University, London, Ontario, Canada
| | - Luis Antonio Diaz
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Cabrera
- Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Escuela de Medicina, Facultad de Ciencias Medicas, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, MASLD Research Center, University of California San Diego, San Diego, California
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
48
|
Marigorta UM, Millet O, Lu SC, Mato JM. Dysfunctional VLDL metabolism in MASLD. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:16. [PMID: 39049993 PMCID: PMC11263124 DOI: 10.1038/s44324-024-00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/22/2024] [Indexed: 07/27/2024]
Abstract
Lipidomics has unveiled the intricate human lipidome, emphasizing the extensive diversity within lipid classes in mammalian tissues critical for cellular functions. This diversity poses a challenge in maintaining a delicate balance between adaptability to recurring physiological changes and overall stability. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), linked to factors such as obesity and diabetes, stems from a compromise in the structural and functional stability of the liver within the complexities of lipid metabolism. This compromise inaccurately senses an increase in energy status, such as during fasting-feeding cycles or an upsurge in lipogenesis. Serum lipidomic studies have delineated three distinct metabolic phenotypes, or "metabotypes" in MASLD. MASLD-A is characterized by lower very low-density lipoprotein (VLDL) secretion and triglyceride (TG) levels, associated with a reduced risk of cardiovascular disease (CVD). In contrast, MASLD-C exhibits increased VLDL secretion and TG levels, correlating with elevated CVD risk. An intermediate subtype, with a blend of features, is designated as the MASLD-B metabotype. In this perspective, we examine into recent findings that show the multifaceted regulation of VLDL secretion by S-adenosylmethionine, the primary cellular methyl donor. Furthermore, we explore the differential CVD and hepatic cancer risk across MASLD metabotypes and discuss the context and potential paths forward to gear the findings from genetic studies towards a better understanding of the observed heterogeneity in MASLD.
Collapse
Affiliation(s)
- Urko M. Marigorta
- Integrative Genomics Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - José M. Mato
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain
| |
Collapse
|
49
|
Reis-Costa A, Belew GD, Viegas I, Tavares LC, Meneses MJ, Patrício B, Gastaldelli A, Macedo MP, Jones JG. The Effects of Long-Term High Fat and/or High Sugar Feeding on Sources of Postprandial Hepatic Glycogen and Triglyceride Synthesis in Mice. Nutrients 2024; 16:2186. [PMID: 39064628 PMCID: PMC11279633 DOI: 10.3390/nu16142186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/03/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND In MASLD (formerly called NAFLD) mouse models, oversupply of dietary fat and sugar is more lipogenic than either nutrient alone. Fatty acids suppress de novo lipogenesis (DNL) from sugars, while DNL inhibits fatty acid oxidation. How such factors interact to impact hepatic triglyceride levels are incompletely understood. METHODS Using deuterated water, we measured DNL in mice fed 18-weeks with standard chow (SC), SC supplemented with 55/45-fructose/glucose in the drinking water at 30% (w/v) (HS), high-fat chow (HF), and HF with HS supplementation (HFHS). Liver glycogen levels and its sources were also measured. For HS and HFHS mice, pentose phosphate (PP) fluxes and fructose contributions to DNL and glycogen were measured using [U-13C]fructose. RESULTS The lipogenic diets caused significantly higher liver triglyceride levels compared to SC. DNL rates were suppressed in HF compared to SC and were partially restored in HFHS but supplied a minority of the additional triglyceride in HFHS compared to HF. Fructose contributed a significantly greater fraction of newly synthesized saturated fatty acids compared to oleic acid in both HS and HFHS. Glycogen levels were not different between diets, but significant differences in Direct and Indirect pathway contributions to glycogen synthesis were found. PP fluxes were similar in HS and HFHS mice and were insufficient to account for DNL reducing equivalents. CONCLUSIONS Despite amplifying the lipogenic effects of fat, the fact that sugar-activated DNL per se barely contributes suggests that its role is likely more relevant in the inhibition of fatty acid oxidation. Fructose promotes lipogenesis of saturated over unsaturated fatty acids and contributes to maintenance of glycogen levels. PP fluxes associated with sugar conversion to fat account for a minor fraction of DNL reducing equivalents.
Collapse
Affiliation(s)
- Ana Reis-Costa
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (A.R.-C.); (G.D.B.)
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Grupo de Estudos de Investigação Fundamental e Translacional (GIFT) da Sociedade Portuguesa de Diabetologia, 1250-198 Lisboa, Portugal
| | - Getachew D. Belew
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (A.R.-C.); (G.D.B.)
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Ivan Viegas
- Centre for Functional Ecology (CFE), TERRA Associate Laboratory, Department of Life Sciences, University of Coimbra, 3030-790 Coimbra, Portugal;
| | - Ludgero C. Tavares
- Vasco da Gama Research Center (CIVG), University School Vasco da Gama, 3020-210 Coimbra, Portugal;
| | - Maria João Meneses
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (M.J.M.); (B.P.); (M.P.M.)
| | - Bárbara Patrício
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (M.J.M.); (B.P.); (M.P.M.)
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 56124 Pisa, Italy;
- Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Amalia Gastaldelli
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 56124 Pisa, Italy;
- Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (M.J.M.); (B.P.); (M.P.M.)
- APDP-Diabetes Portugal Education and Research Center (APDP-ERC), 1250-203 Lisboa, Portugal
| | - John G. Jones
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Grupo de Estudos de Investigação Fundamental e Translacional (GIFT) da Sociedade Portuguesa de Diabetologia, 1250-198 Lisboa, Portugal
| |
Collapse
|
50
|
Barrera F, Uribe J, Olvares N, Huerta P, Cabrera D, Romero-Gómez M. The Janus of a disease: Diabetes and metabolic dysfunction-associated fatty liver disease. Ann Hepatol 2024; 29:101501. [PMID: 38631419 DOI: 10.1016/j.aohep.2024.101501] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2024]
Abstract
Metabolic Dysfunction-Associated Fatty Liver Disease and Diabetes Mellitus are two prevalent metabolic disorders that often coexist and synergistically contribute to the progression of each other. Several pathophysiological pathways are involved in the association, including insulin resistance, inflammation, and lipotoxicity, providing a foundation for understanding the complex interrelationships between these conditions. The presence of MASLD has a significant impact on diabetes risk and the development of microvascular and macrovascular complications, and diabetes significantly contributes to an increased risk of liver fibrosis progression in MASLD and the development of hepatocellular carcinoma. Moreover, both pathologies have a synergistic effect on cardiovascular events and mortality. Therapeutic interventions targeting MASLD and diabetes are discussed, considering lifestyle modifications, pharmacological agents, and emerging treatment modalities. The review also addresses the challenges in managing these comorbidities, such as the need for personalized approaches and the potential impact on cardiovascular health. The insights gleaned from this analysis can inform clinicians, researchers, and policymakers in developing integrated strategies for preventing, diagnosing, and managing these metabolic disorders.
Collapse
Affiliation(s)
- Francisco Barrera
- Laboratorio Experimental de Hepatología, Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Javier Uribe
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nixa Olvares
- Laboratorio Experimental de Hepatología, Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Programa de Immunogenética e Inmunología traslacional, Instituto de Ciencias e Inovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Paula Huerta
- Programa de Medicina Interna, Instituto de Ciencias e Inovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile; Hospital Padre Hurtado, Santiago, Chile
| | - Daniel Cabrera
- Laboratorio Experimental de Hepatología, Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Escuela de Medicina, Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago, Chile
| | - Manuel Romero-Gómez
- Enfermedades Digestivas y Ciberehd, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (CSIC/HUVR/US), Universidad de Sevilla, Sevilla, España.
| |
Collapse
|