1
|
Oğuz E, Yılmaz Y, Güneş FE. The relationship between bacterial changes and dietary intervention in non-alcoholic fatty liver disease. Clin Nutr ESPEN 2025; 68:267-273. [PMID: 40345652 DOI: 10.1016/j.clnesp.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/19/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND AND AIM This study aimed to investigate the levels of Faecalibacterium prausnitzii (F. prausnitzii) and Eubacterium rectale (E. rectale) in patients with non-alcoholic fatty liver disease (NAFLD) and evaluate the impact of dietary intervention on these bacterial populations. MATERIALS An interventional study was conducted with 38 NAFLD patients recruited from the Marmara University Gastroenterology Institute. Participants were divided into two groups: a diet intervention group (n = 21) and a control group (n = 17). The dietary intervention consisted of a balanced diet containing 50-55 % carbohydrates, 30-35 % fats, and 15-20 % protein. Fecal samples were collected at baseline and after six weeks for both groups, and bacterial quantification was performed via deoxyribonucleic acid (DNA) analysis of the fecal samples. RESULTS In the diet intervention group, a significant increase in E. rectale abundance was observed after six weeks (p = 0.008). Additionally, intakes of dietary fiber, vitamin E, vitamin C, and thiamine were significantly higher in the intervention group compared to the control group by the end of the study (p < 0.05). However, no significant changes were detected in F. prausnitzii levels in either group. CONCLUSION The findings demonstrate that dietary intervention can significantly increase E. rectale abundance in NAFLD patients, while F. prausnitzii levels remain unaffected. These results highlight the selective influence of dietary modifications on gut bacterial populations, offering potential implications for the management of NAFLD.
Collapse
Affiliation(s)
- Esma Oğuz
- Kırklareli University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Kırklareli, Türkiye.
| | - Yusuf Yılmaz
- Recep Tayyip Erdoğan University, School of Medicine, Department of Gastroenterology, Rize, Türkiye
| | - Fatma Esra Güneş
- İstanbul Medeniyet University, Faculty of Health Sciences, Department of Nutrition and Dietetics, İstanbul, Türkiye
| |
Collapse
|
2
|
Mailloux RJ. Targeted Redox Regulation α-Ketoglutarate Dehydrogenase Complex for the Treatment of Human Diseases. Cells 2025; 14:653. [PMID: 40358176 PMCID: PMC12071522 DOI: 10.3390/cells14090653] [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: 04/09/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
α-ketoglutarate dehydrogenase complex (KGDHc) is a crucial enzyme in the tricarboxylic acid (TCA) cycle that intersects monosaccharides, amino acids, and fatty acid catabolism with oxidative phosphorylation (OxPhos). A key feature of KGDHc is its ability to sense changes in the redox environment through the reversible oxidation of the vicinal lipoic acid thiols of its dihydrolipoamide succinyltransferase (DLST; E2) subunit, which controls its activity and, by extension, OxPhos. This characteristic inculcates KGDHc with redox regulatory properties for the modulation of metabolism and mediating of intra- and intercellular signals. The innate capacity of KGDHc to participate in the regulation of cell redox homeodynamics also occurs through the production of mitochondrial hydrogen peroxide (mtH2O2), which is generated by the dihydrolipoamide dehydrogenase (DLD; E3) downstream from the E2 subunit. Reversible covalent redox modification of the E2 subunit controls this mtH2O2 production by KGDHc, which not only protects from oxidative distress but also modulates oxidative eustress pathways. The importance of KGDHc in modulating redox homeodynamics is underscored by the pathogenesis of neurological and metabolic disorders that occur due to the hyper-generation of mtH2O2 by this enzyme complex. This also implies that the targeted redox modification of the E2 subunit could be a potential therapeutic strategy for limiting the oxidative distress triggered by KGDHc mtH2O2 hyper-generation. In this short article, I will discuss recent findings demonstrating KGDHc is a potent mtH2O2 source that can trigger the manifestation of several neurological and metabolic diseases, including non-alcoholic fatty liver disease (NAFLD), inflammation, and cancer, and the targeted redox modification of the E2 subunit could alleviate these syndromes.
Collapse
Affiliation(s)
- Ryan J Mailloux
- School of Human Nutrition, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Quebec, QC H9X 3V9, Canada
| |
Collapse
|
3
|
Jiang M, Butt AS, Cua IH, Pan Z, Al-Busafi SA, Méndez-Sánchez N, Eslam M. MAFLD vs. MASLD: a year in review. Expert Rev Endocrinol Metab 2025:1-12. [PMID: 40237514 DOI: 10.1080/17446651.2025.2492767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
INTRODUCTION In 2023, metabolic dysfunction-associated steatotic liver disease (MASLD) was introduced following metabolic dysfunction-associated fatty liver disease (MAFLD). Both aim to address the limitations of nonalcoholic fatty liver disease (NAFLD). This review analyzes the similarities and differences between MAFLD and MASLD, focusing on their impacts on epidemiology, diagnosis, stigma, and related liver diseases. AREAS COVERED Current evidence suggests that MAFLD criteria effectively identify individuals at higher risk through a good balance of sensitivity and specificity. Moreover, MAFLD is a more generalizable term that is easily understood globally. EXPERT OPINION The transition from NAFLD to MAFLD and MASLD marks a significant advance in understanding fatty liver disease within hepatology. MAFLD identifies a homogeneous cohort of patients with fatty liver due to metabolic dysfunction and provides a valuable framework for holistic, patient-centered management strategies that consider various contributing factors to improve health outcomes.
Collapse
Affiliation(s)
- Mingqian Jiang
- Department of Endocrinology and Metabolism, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia
| | - Amna Subhan Butt
- Section of Gastroenterology, Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - Ian Homer Cua
- Institute of Digestive and Liver Diseases, St. Luke's Medical Center, Taguig, Philippines
| | - Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia
| | - Said A Al-Busafi
- Gastroenterology and Hepatology Unit, Department of Medicine, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic and Foundation, National Autonomous University of Mexico, Mexico City, Mexico
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
4
|
Milani I, Chinucci M, Leonetti F, Capoccia D. MASLD: Prevalence, Mechanisms, and Sex-Based Therapies in Postmenopausal Women. Biomedicines 2025; 13:855. [PMID: 40299427 PMCID: PMC12024897 DOI: 10.3390/biomedicines13040855] [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: 03/08/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease influenced by genetic, lifestyle, and environmental factors. While MASLD is more prevalent in men, women are at increased risk after menopause, highlighting the critical pathogenetic role of sex hormones. The complex interplay between estrogen deficiency, visceral fat accumulation, metabolic syndrome (MetS), and inflammation accelerates disease progression, increases cardiovascular (CV) risk, and triggers a cycle of worsening adiposity, metabolic dysfunction, and psychological problems, including eating disorders. Weight loss in postmenopausal women can significantly improve both metabolic and psychological outcomes, helping to prevent MASLD and related conditions. This review examines the prevalence of MASLD, its comorbidities (type 2 diabetes T2D, CV, mental disorders), pathogenetic mechanisms, and pharmacological treatment with GLP-1 receptor agonists (GLP1-RAs), with a focus on postmenopausal women. Given the use of GLP1-RAs in the treatment of obesity and T2D in MASLD patients, and the increase in MetS and MASLD after menopause, this review analyzes the potential of a stable GLP-1-estrogen conjugate as a therapeutic approach in this subgroup. By combining the synergistic effects of both hormones, this dual agonist has been shown to increase food intake and food reward suppression, resulting in greater weight loss and improved insulin sensitivity, glucose, and lipid metabolism. Therefore, we hypothesize that this pharmacotherapy may provide more targeted therapeutic benefits than either hormone alone by protecting the liver, β-cells, and overall metabolic health. As these effects are only supported by preclinical data, this review highlights the critical need for future research to evaluate and confirm the mechanisms and efficacy in clinical settings, particularly in postmenopausal women.
Collapse
Affiliation(s)
- Ilaria Milani
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (M.C.); (F.L.); (D.C.)
| | | | | | | |
Collapse
|
5
|
Souza M, Al-Sharif L, Diaz I, Khalil SM, Lv XH, Mantovani A, Villela-Nogueira CA. Representation of Sex, Race and Ethnicity in MASH Randomised Controlled Trials: A Systematic Review and Meta-Analysis. Liver Int 2025; 45:e70029. [PMID: 40029148 DOI: 10.1111/liv.70029] [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: 10/24/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND AND AIMS Randomised controlled trials (RCTs) have historically underrepresented female, racial and ethnic minorities across various fields. This systematic review and meta-analysis aims to examine the global distribution, reporting and participation of diverse groups based on sex, race and ethnicity in trials focused on metabolic dysfunction-associated steatohepatitis (MASH). METHODS PubMed and Cochrane Library databases were systematically searched for MASH RCTs (through December 13, 2024) that included any pharmacotherapy as an intervention arm. RCTs were qualitatively reviewed to assess their global distribution and reporting of populations. A meta-analysis of proportions was performed using a generalised linear mixed model. RESULTS One hudred and nine studies were identified, reporting data from 112 RCTs and 19 516 MASH participants. Of the 49 countries that conducted trials, 34 were high-income countries (69.4%). Sex, race and ethnicity were reported in 111 (99.1%), 69 (61.6%) and 56 (50.0%) of the 112 RCTs, respectively, with reporting improving in recent years. We found no reporting of sexual and gender minorities. The pooled proportions of female, White, Asian, Black and Hispanic/Latino groups were 54.23% (95% confidence interval [CI]: 51.31-57.12), 87.63% (95% CI: 85.37-89.58), 4.95% (95% CI: 3.42-7.10), 2.27% (95% CI: 1.89-2.71) and 31.42% (95% CI: 26.61-36.66), respectively. Meta-regressions showed a trend toward more female, White and Hispanic/Latino participants in RCTs over time. CONCLUSIONS Although female and Hispanic/Latino representation has increased over time, racial minorities are underrepresented in MASH trials. These data provide an overview of participant representation in MASH trials and call for collaborative efforts among researchers, sponsors, regulators and other relevant stakeholders to improve diversity in these trials.
Collapse
Affiliation(s)
- Matheus Souza
- Department of Internal Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lubna Al-Sharif
- Department of Biomedical Sciences and Basic Clinical Skills, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Ivanna Diaz
- Department of Internal Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | | | - Xiu-He Lv
- Department of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | | |
Collapse
|
6
|
Xu K, Corona-Avila I, Frutos MD, Núñez-Sánchez MÁ, Makhanasa D, Shah PV, Guzman G, Ramos-Molina B, Priyadarshini M, Khan MW. Hepatic HKDC1 deletion alleviates western diet-induced MASH in mice. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167746. [PMID: 40020530 DOI: 10.1016/j.bbadis.2025.167746] [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: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
The global prevalence of Metabolic Dysfunction-Associated Steatohepatitis (MASH) has been rising sharply, closely mirroring the increasing rates of obesity and metabolic syndrome. MASH exhibits a strong sexual dimorphism where females are affected with more severe forms after menopause. Hexokinase domain-containing protein 1 (HKDC1) has recently been recognized for its role in liver diseases, where its expression is minimal under normal conditions but significantly increases in response to metabolic stressors like obesity and liver injury. This selective upregulation suggests HKDC1's potential specialization in hepatic glucose and lipid dysregulation, linking it closely to the progression of MASH. This study aims to clarify the role of HKDC1 in Western diet-induced MASH in female mice by examining its impact on hepatic glucose and lipid metabolism, offering insights into its potential as a therapeutic target and addressing the need for sex-specific research in liver disease. This study reveals that HKDC1 expression is elevated in obese women with MASH and correlates with liver pathology. In a mouse model, liver-specific HKDC1 knockout (HKDC1LKO) protected against Western diet-induced obesity, glucose intolerance, and MASH features, including steatosis, inflammation, and fibrosis. Transcriptomic analysis showed that HKDC1 deletion reduced pro-inflammatory and pro-fibrotic gene expression, while gut microbiome analysis indicated a shift toward MASH-protective bacteria. These findings suggest that HKDC1 may exacerbate MASH progression through its role in metabolic and inflammatory pathways, making it a potential therapeutic target.
Collapse
Affiliation(s)
- Kai Xu
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - Irene Corona-Avila
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, 30120 Murcia, Spain
| | - María Ángeles Núñez-Sánchez
- Obesity, Diabetes and Metabolism Research Group, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain
| | - Dhruvi Makhanasa
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - Pratham Viral Shah
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Research Group, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain
| | - Medha Priyadarshini
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America.
| | - Md Wasim Khan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America.
| |
Collapse
|
7
|
Tamini S, Bondesan A, Caroli D, Marazzi N, Sartorio A. The Ability of the Triglyceride-Glucose (TyG) Index and Modified TyG Indexes to Predict the Presence of Metabolic-Associated Fatty Liver Disease and Metabolic Syndrome in a Pediatric Population with Obesity. J Clin Med 2025; 14:2341. [PMID: 40217790 PMCID: PMC11989838 DOI: 10.3390/jcm14072341] [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: 03/10/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Metabolic-associated fatty liver disease (MASLD) and metabolic syndrome (MetS) are increasingly prevalent among children and adolescents with obesity, posing significant long-term metabolic and cardiovascular risks. Non-invasive identification of at-risk individuals is crucial for a timely intervention. This study aimed to evaluate the diagnostic performance of the triglyceride-glucose (TyG) index and its modified versions, TyG-body mass index (TyG-BMI) and TyG-waist circumference (TyG-WC), in predicting MASLD and MetS in a large cohort of children and adolescents with obesity. Methods: A total of 758 children and adolescents with obesity (454 females, 304 males; mean age 14.8 ± 2.1 years; mean BMI 37.9 ± 6.2 kg/m2) were included. MASLD was diagnosed via ultrasonography, while MetS was defined using International Diabetes Federation criteria. TyG, TyG-WC, and TyG-BMI were calculated for all participants. Receiver operating characteristic (ROC) curves were generated to assess the diagnostic accuracy of these indexes, including sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). Results: MASLD was detected in 38.9% of participants, with a higher prevalence in males (p < 0.0001). MetS was present in 27.8% of the cohort, with higher prevalence in males (p < 0.0001). Among the indexes, TyG-WC exhibited the highest sensitivity for MASLD (77.6%), whereas TyG-BMI had the highest specificity (63.3%). In predicting MetS, all three indexes performed better than for MASLD, with TyG demonstrating the highest PPV (54.5%) and TyG-BMI the highest NPV (87.5%). Predictive performance was lower in males than females, potentially due to sex-specific differences in fat distribution and metabolic response. Conclusions: TyG, TyG-WC, and TyG-BMI are promising, non-invasive tools for identifying children and adolescents with obesity at risk for MASLD and MetS. The superior sensitivity of TyG-WC and the high specificity of TyG-BMI highlight the value of incorporating anthropometric parameters into metabolic screening. Integrating these indexes into routine clinical practice may enhance early detection, allowing for timely intervention and personalized management strategies, ultimately reducing the long-term burden of metabolic and liver diseases in pediatric populations.
Collapse
Affiliation(s)
- Sofia Tamini
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-Endocrinological Research, 28824 Piancavallo-Verbania, Italy; (A.B.); (D.C.); (N.M.); (A.S.)
| | | | | | | | | |
Collapse
|
8
|
Kasarinaite A, Ramos MJ, Beltran-Sierra M, Sutherland EF, Rei PA, Zhao M, Chi Y, Beniazza M, Corsinotti A, Kendall TJ, Henderson NC, Fallowfield JA, Saunders PTK, Hay DC. Hormone correction of dysfunctional metabolic gene expression in stem cell-derived liver tissue. Stem Cell Res Ther 2025; 16:130. [PMID: 40069876 PMCID: PMC11899078 DOI: 10.1186/s13287-025-04238-0] [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: 11/20/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
The increase in metabolic dysfunction-associated steatotic liver disease (MASLD) and its progression to metabolic dysfunction-associated steatohepatitis (MASH) is a worldwide healthcare challenge. Heterogeneity between men and women in the prevalence and mechanisms of MASLD and MASH is related to differential sex hormone signalling within the liver, and declining hormone levels during aging. In this study we used biochemically characterised pluripotent stem cell derived 3D liver spheres to model the protective effects of testosterone and estrogen signalling on metabolic liver disease 'in the dish'. We identified sex steroid-dependent changes in gene expression which were protective against metabolic dysfunction, fibrosis, and advanced cirrhosis patterns of gene expression, providing new insight into the pathogenesis of MASLD and MASH, and highlighting new druggable targets. Additionally, we highlight gene targets for which drugs already exist for future translational studies.
Collapse
Affiliation(s)
- Alvile Kasarinaite
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Maria Jimenez Ramos
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Mariana Beltran-Sierra
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Elena F Sutherland
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Pedro Arede Rei
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Make Zhao
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University, Haining, China
| | - Ying Chi
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University, Haining, China
| | - Meryam Beniazza
- Single-Cell Multi-Omics Facility, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Andrea Corsinotti
- Single-Cell Multi-Omics Facility, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Timothy J Kendall
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jonathan A Fallowfield
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - Philippa T K Saunders
- Centre for Reproductive Health, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK
| | - David C Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4UU, UK.
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University, Haining, China.
| |
Collapse
|
9
|
Barr B, Levitt DE, Gollahon L. Red Meat Amino Acids for Beginners: A Narrative Review. Nutrients 2025; 17:939. [PMID: 40289994 PMCID: PMC11946737 DOI: 10.3390/nu17060939] [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: 02/19/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/30/2025] Open
Abstract
Meat is a major source of dietary protein and fat across the globe. Red and white meat are the major terms consumers use to refer to types of meat; however, these terms do not fully encompass the range of nutrients provided by meat sources. Red meat refers to meat from mammalian skeletal muscle, while white meat refers to poultry. Red and white meat both provide a wide range of nutritional components in the context of fatty acids, amino acids and micronutrients. Importantly, it has been demonstrated that amino acid profiles differ between red meat and white meat as well as between different sources of red meat. Red meat is a complete source of dietary amino acids, meaning it contains all essential amino acids (EAAs), and in addition, it contains all the non-essential amino acids (NEAAs). Red meat is also the most abundant source of bioavailable heme-iron essential for muscle growth and cardiovascular health. Red meat has been indicated as a major contributor to the rising incidence of metabolic disorders and even colorectal cancer. However, it is important to note that while red meat consumption is linked to these conditions, it is typically the overconsumption of red meat that is associated with obesity and other metabolic symptoms. Similarly, the preparation of red meat is a key factor in its link to colorectal cancer as some methods of preparation produce carcinogens while others do not. Finally, red meat may also be situationally more beneficial to some groups than others, particularly in the cases of sex and aging. For pregnant women, increases in red meat consumption may be beneficial to increase the intake of semi-essential amino acids, while in the elderly, increases in red meat consumption may better preserve muscle mass compared with other dietary protein sources.
Collapse
Affiliation(s)
- Benjamin Barr
- Department of Biological Sciences, Texas Tech University, 2500 Main Street, Lubbock, TX 79409, USA;
| | - Danielle E. Levitt
- Department of Kinesiology and Sports Management, Texas Tech University, 3204 Main Street, Lubbock, TX 79409, USA;
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Main Street, Lubbock, TX 79409, USA;
| |
Collapse
|
10
|
Kim S, Yoo HY. Sex differences in predicting dyslipidemia using polygenic risk score with fatty liver index and fibrotic nonalcoholic steatohepatitis index. Sci Rep 2025; 15:7849. [PMID: 40050666 PMCID: PMC11885555 DOI: 10.1038/s41598-025-92766-3] [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: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 03/09/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) are recognized risk factors for dyslipidemia. Current prediction models that rely solely on dyslipidemia polygenic risk score (PRS) have certain limitations. We aimed to validate simple indexes for NAFLD and NASH as predictors of dyslipidemia using the PRS. This study utilized cohort data from an urban population-based dataset comprising 48,263 South Koreans. The incidence of dyslipidemia was higher in men than in women (32.4% and 27.8%; p < 0.001). The PRS model predicted dyslipidemia more accurately in men (AUROC [95% confidence intervals]: 0.645 [0.636-0.754]). Notably, integrating the fatty liver index (FLI) and fibrotic NASH index (FNI) with the PRS model resulted in the highest accuracy in diagnosing dyslipidemia, particularly in men (AUROC [95% confidence intervals]: 0.704 [0.698-0.711]). In conclusion, a predictive model combining the PRS with FLI and FNI was validated. This model offers more accurate predictive value for diagnosing dyslipidemia, particularly in East Asian men. Thus, our study has the clinical potential for identifying high-risk individuals and determining preventive measures for dyslipidemia in a sex-specific manner.
Collapse
Affiliation(s)
- Sei Kim
- Department of Nursing, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hae Young Yoo
- Department of Nursing, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
11
|
Zhang Q, Wang Y, Liu S, Zhu S, Li P, Wu S. Mortality risk associated with MASLD, MASLD type and different cardiometabolic risk factors in IBD patients: A long-term prospective cohort study. Dig Liver Dis 2025; 57:744-752. [PMID: 39581836 DOI: 10.1016/j.dld.2024.11.002] [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: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE To examine the mortality risk associated with metabolic dysfunction-associated steatotic liver disease (MASLD), MASLD type, lean/non-lean MASLD and different cardiometabolic risk factors (CMRFs) in patients with inflammatory bowel disease (IBD) based on a long-term prospective cohort. METHODS Prevalent IBD patients at baseline who were free of alcoholic liver disease, cancer and hepatitis B/C virus seropositive were included (N=4622). MASLD, MASLD type [pure MASLD, MASLD with increased alcohol intake (MetALD)], lean/non-lean MASLD and CMRFs at baseline were defined according to the latest criteria proposed by AASLD and EASL. Primary outcome was all-cause mortality. Cox proportional hazard model was used to examine the association. RESULTS Overall, 1,763 (38.1%) were diagnosed with MASLD. During a median of 13.3-year follow-up, 451 all-cause deaths were identified. Compared with IBD-only patients, those with MASLD had a 58% excess risk of mortality (HR=1.58, 95%CI:1.07-2.32). Furthermore, as number of CMRFs increased in MASLD patients, mortality risk was significantly increased (Ptrend=0.005), with a 85% and 83% higher risk in MASLD with 3 CMRFs (HR=1.85, 95%CI:1.20-2.85) and ≥4 CMRFs (HR=1.83, 95%CI:1.16-2.89) versus IBD-only patients. Specifically, similar elevated mortality risk was observed in either pure MASLD (HR= 1.62, 95%CI:1.09-2.43) or MetALD (HR=2.03, 95%CI:1.24-3.32). Moreover, the excess mortality risk was both indicated in lean (HR=3.14, 95%CI:1.57-6.29) and non-lean MASLD (HR=1.67, 95%CI:1.12-2.48). CONCLUSIONS MASLD, either pure MASLD or MetALD, as well as lean/non-lean MASLD, is associated with increased mortality risk in IBD patients, with greater risk as number of cardiometabolic risk factors increased and evidently higher risk in lean MASLD patients.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Yutao Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Shanshan Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University; State Key Laboratory of Digestive Health; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
12
|
Li Y, Fan W, Lo TH, Jiang JX, Fish SR, Tomilov A, Chronopoulos A, Bansal V, Mozes G, Vancza L, Kunimoto K, Ye J, Becker L, Das S, Park H, Wei Y, Ranjbarvaziri S, Bernstein D, Ramsey J, Cortopassi G, Török NJ. P46Shc Inhibits Mitochondrial ACAA2 Thiolase, Exacerbating Mitochondrial Injury and Inflammation in Aging Livers. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:528-541. [PMID: 39733992 PMCID: PMC11841488 DOI: 10.1016/j.ajpath.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/08/2024] [Accepted: 10/29/2024] [Indexed: 12/31/2024]
Abstract
Mitochondrial maladaptation and dysfunction contribute to the progression of metabolic dysfunction-associated steatohepatitis (MASH). Induction of Shc is implicated in progressive MASH during aging and the cytoplasmic p52Shc isoform in the activation of redox enzyme NOX2. The mitochondrial Shc isoform p46Shc represses acetyl-coenzyme A acyltransferase 2 (ACAA2) in vitro. ACAA2 is a key enzyme for lipid β-oxidation; however, the metabolic consequences of in vivo p46Shc induction are unknown. In the current study, p46Shc-inducible mice were generated; these and littermate controls were aged and fed chow or fast-food (high-fat and high-fructose) diet. p46Shc induction increased liver injury, inflammation, and lipid peroxidation. p46Shc overexpression did not significantly change liver triglycerides. On electron microscopy studies, mitochondria were swollen with aberrant cristae. p46Shc induction reduced mitochondrial oxygen consumption as measured by Oroboros, as well as suppressed the production of β-hydroxybutyrate, the central metabolite of therapeutic ketosis. Mitochondria exhibited increased production of reactive oxidative species. By contrast, the expression of dominant negative p46Shc reduced ACAA2 thiolase activity, improved β-oxidation, and reduced lipid peroxidation and production of reactive oxidative species. In summary, these studies support the concept that p46Shc induction in aging represses ACAA2, resulting in decreased mitochondrial β-oxidation and increased lipid peroxidation. Maintaining β-oxidation and ketogenesis could prevent liver injury, and targeting Shc-related maladaptive responses could be a successful therapeutic strategy in aging/MASH.
Collapse
Affiliation(s)
- Yuan Li
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Weiguo Fan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Tzu-Han Lo
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Joy X Jiang
- Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, California
| | - Sarah R Fish
- Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, California
| | - Alexey Tomilov
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, Davis, California
| | - Antonios Chronopoulos
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Vidushi Bansal
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Gergely Mozes
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Lorand Vancza
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Koshi Kunimoto
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Jiayu Ye
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Laren Becker
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Suvarthi Das
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Hyesuk Park
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Yi Wei
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California
| | - Sara Ranjbarvaziri
- Department of Pediatrics, Stanford University, Stanford, California; Cardiovascular Research Institute, Stanford University, Stanford, California
| | - Daniel Bernstein
- Department of Pediatrics, Stanford University, Stanford, California; Cardiovascular Research Institute, Stanford University, Stanford, California
| | - Jon Ramsey
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, Davis, California
| | - Gino Cortopassi
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, Davis, California
| | - Natalie J Török
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California; Palo Alto VA Medical Center, Palo Alto, California.
| |
Collapse
|
13
|
Higa R, Pourteymour S, Kolan PS, Dankel SN, Fernø J, Mellgren G, Pan C, Seldin MM, Lusis AJ, Drevon CA, Dalen KT, Norheim FA. Hepatic lipid metabolism is altered in Ubiad1 +/- mice of both sexes. Sci Rep 2025; 15:7022. [PMID: 40016272 PMCID: PMC11868635 DOI: 10.1038/s41598-025-91283-7] [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: 05/13/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
UbiA prenyltransferase domain containing 1 (Ubiad1) has the potential to affect cholesterol and phospholipid levels in different cell types. We previously identified Ubiad1 as a candidate gene for regulating subcutaneous fat pad weight in a mouse genome-wide association study. Here we evaluated the relationship between Ubiad1 and obesity-related traits in cohorts of humans and mice, and in Ubiad1+/- mice fed a high-fat diet. In both humans and mice, adipose tissue Ubiad1 mRNA expression correlated negatively with adiposity and positively with mitochondria-related genes. To determine the role of Ubiad1 in high-fat diet-induced obesity, we disrupted the Ubiad1 gene in mice. Deletion of Ubiad1 was embryonically lethal in C57BL/6 N mice, preventing analysis of adult Ubiad1-/- mice. Thus, male and female Ubiad1+/+ and Ubiad1+/- mice were fed high-fat diet for 10 weeks, with no difference in weight gain and adipose tissue organ weights observed between the genotypes. Analysis of liver mRNA expression revealed that Ubiad1 heterozygosis (Ubiad1+/-) altered several pathways involved in lipid metabolism. Detailed lipid quantification with HPLC-qTOF/MS showed increased levels of hepatic ceramides in female Ubiad1+/- mice, whereas phosphatidylglycerols, phosohatidylinositol and lysophosphatidylethanolamines were reduced in male Ubiad1+/- mice. Our findings reveal sex-specific effects of Ubiad1 expression on hepatic lipid metabolism.
Collapse
Affiliation(s)
- Ryoko Higa
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Shirin Pourteymour
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Pratibha S Kolan
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Simon N Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Johan Fernø
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, University of California, 650 Charles E Young Drive South, Los Angeles, Los Angeles, CA, 90095, USA
- Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, University of California, los Angeles, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, University of California, 650 Charles E Young Drive South, Los Angeles, Los Angeles, CA, 90095, USA
- Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, University of California, los Angeles, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
- Vitas AS, Science Park, Gaustadalléen 21, 0349, Oslo, Norge
| | - Knut T Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Frode A Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway.
| |
Collapse
|
14
|
Xu K, Corona-Avila I, Frutos MD, Nunez-Sanchez MA, Makhanasa D, Shah PV, Guzman G, Ramos-Molina B, Priyadarshini M, Khan MW. Hepatic HKDC1 Deletion Alleviates Western Diet-Induced MASH in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625530. [PMID: 39651120 PMCID: PMC11623584 DOI: 10.1101/2024.11.26.625530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The global prevalence of Metabolic dysfunction-associated steatohepatitis (MASH) has been rising sharply, closely mirroring the increasing rates of obesity and metabolic syndrome. MASH exhibits a strong sexual dimorphism where females are affected with more severe forms after menopause. Hexokinase domain-containing protein 1 (HKDC1) has recently been recognized for its role in liver diseases, where its expression is minimal under normal conditions but significantly increases in response to metabolic stressors like obesity and liver injury. This selective upregulation suggests HKDC1s potential specialization in hepatic glucose and lipid dysregulation, linking it closely to the progression of MASLD and MASH. This study aims to clarify the role of HKDC1 in Western diet-induced MASH in female mice by examining its impact on hepatic glucose and lipid metabolism, offering insights into its potential as a therapeutic target and addressing the need for sex-specific research in liver disease. This study reveals that HKDC1 expression is elevated in obese women with MASH and correlates with liver pathology. In a mouse model, liver-specific HKDC1 knockout (HKDC1LKO) protected against Western diet-induced obesity, glucose intolerance, and MASH features, including steatosis, inflammation, and fibrosis. Transcriptomic analysis showed that HKDC1 deletion reduced pro-inflammatory and pro-fibrotic gene expression, while gut microbiome analysis indicated a shift toward MASH-protective bacteria. These findings suggest that HKDC1 may exacerbate MASH progression through its role in metabolic and inflammatory pathways, making it a potential therapeutic target.
Collapse
|
15
|
Nilghaz M, Sadeghi A, Koochakpoor G, Poustchi H, Khodadadi N, Narimani B, Ghods M, Shafiee M, Shahparvari MR, Hekmatdoost A. The efficacy of DASH combined with time-restricted feeding (16/8) on metabolic associated fatty liver disease management: a randomized controlled trial. Sci Rep 2025; 15:7020. [PMID: 40016311 PMCID: PMC11868424 DOI: 10.1038/s41598-025-88393-7] [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: 07/14/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Recent studies have utilized time-restricted feeding (16/8) (TRF) and dietary approaches to stop hypertension separately to manage metabolic-associated fatty liver disease (MAFLD); however, the effectiveness of combining these two approaches has not been investigated. The objective of this study was to examine the impact of TRF in conjunction with a DASH diet on various factors related to MAFLD. A 12-week randomized controlled trial was conducted to assess the impact of TRF (16/8), along with a DASH diet, compared with a control diet based on standard meal distribution, in patients with MAFLD. An investigation was conducted to examine alterations in anthropometric indices, as well as liver parameters, serum metabolic indices, and an inflammatory marker. The TRF plus DASH diet reduced body mass index (p = 0.03), abdominal circumference (p = 0.005), controlled attenuation parameter (CAP) (p < 0.001), alanine aminotransferase (p = 0.039), and aspartate aminotransferase (0.047) compared to the control group. The levels of insulin and homeostasis model assessment of insulin resistance reduced in both groups significantly (P < 0.05). In MAFLD patients, TRF (16/8) in combination with a DASH diet is superior to a low-calorie diet in promoting obesity indices, and hepatic steatosis and fibrosis. Further long-term investigations are needed to draw definitive conclusions.
Collapse
Affiliation(s)
- Maryam Nilghaz
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Hossein Poustchi
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Navideh Khodadadi
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnaz Narimani
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Ghods
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshad Shafiee
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Shahparvari
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition, School of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Shu Q, Zhu J, Mo J, Wei X, Zhu Z, Chen X, He F, Zhong L. Identification and validation of PANoptosis-related LncRNAs prognosis system in hepatocellular carcinoma. Sci Rep 2025; 15:6030. [PMID: 39972122 PMCID: PMC11840146 DOI: 10.1038/s41598-025-90498-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] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common solid malignancies in the world. Due to the limited effectiveness of current drug treatments, further research on HCC is necessary. PANoptosis is defined as an inflammatory RCD whose main features combine pyroptosis, apoptosis and necroptosis which cannot be explained by any of these three RCDs alone. In HCC, risk stratification based on PANoptosis-associated lncRNAs has clinical application potential. In this study, we explored HCC related PANoptosis-related lncRNAs (PRLs) by analyzing significantly differentially expressed genes in HCC. HCC-associated PRL scores were established by WGCNA, LASSO analysis and multivariate Cox assessment. Subsequently, we verified the prognostic analysis ability of PRL score for HCC patients, and on this basis established a prognostic risk assessment model for HCC and verified its reliability. The relationship between PRL score and immune infiltration as well as drug sensitivity was further analyzed to evaluate the clinical reference value of this model. Western blot analysis and PCR further verified the reliability of bioinformatics results. The observed suppression of HCC progression and invasiveness following selected PRL knockdown further validated the reliability of our bioinformatics analysis results. Our results provide new evidence for the role of PANoptosis-associated lncRNAs in HCC.
Collapse
Affiliation(s)
- Qi Shu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Junfeng Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jiaping Mo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xiaoyan Wei
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zhenjie Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Xiaojuan Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Fugen He
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Like Zhong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
17
|
Zhao Y, Li J, Ma A, Wang Z, Ni Y, Wu D, Zhou Y, Zhang N, Zhang L, Chang Y, Wang Q. Irisin alleviates hepatic steatosis by activating the autophagic SIRT3 pathway. Chin Med J (Engl) 2025:00029330-990000000-01430. [PMID: 39965865 DOI: 10.1097/cm9.0000000000003427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Disruption of hepatic lipid homeostasis leads to excessive hepatic triglyceride accumulation and the development of metabolic dysfunction-associated steatotic liver disease (MASLD). Autophagy, a critical process in liver lipid metabolism, is impaired in MASLD pathogenesis. Irisin, a skeletal muscle-driven myokine, regulates lipid metabolism, but its impact on hepatic lipid metabolism is not well understood. Here, we aimed to explore the role of irisin in hepatic steatosis and the underlying mechanisms involved. METHODS A high-fat diet (HFD)-induced MASLD mouse model was used, and the recombinant irisin protein, herein referred to as "Irisin", was intraperitoneally administered for 4 weeks to evaluate the effects of irisin on hepatic lipid accumulation. Liver tissues were stained with Oil red O (ORO), and triglyceride (TG) and total cholesterol (TC) contents were measured in serum and liver homogenates. The expression of the autophagosome marker microtubule-associated protein 1 light chain 3 (LC3), the autophagy receptor protein sequestosome-1 (SQSTM1/p62), autophagy initiation complex unc-51-like kinase 1 (ULK1) and the lysosomal functional protein cathepsin B was measured via Western blotting, and the expression of the transcription factor EB (TFEB) was analyzed via immunofluorescence to explore autophagic changes. The effect of irisin on autophagic flux was further evaluated in palmitic acid-induced HepG2 cells by measuring autophagic degradation with chloroquine (CQ), and analyzing the colocalization of LC3 and lysosome-associated protein 1 (LAMP1). The possible mechanism was examined by measuring the expression of the autophagic sirtuin 3 (SIRT3) pathway and further validated using overexpression of SIRT3 with plasmid transfection or siRNA-mediated knockdown. Student's t-test was utilized for statistical analysis. RESULTS Irisin significantly reduces hepatic lipid accumulation in mice fed with HFD, accompanied by enhanced hepatocyte autophagy and upregulation of the SIRT3 pathway. In HepG2 cells, Irisin attenuated palmitic acid-induced lipid accumulation, which was partially dependent on SIRT3 levels. Mechanistically, Irisin treatment upregulated SIRT3 and phosphorylated AMP-activated protein kinase (AMPK), inhibited mammalian target of rapamycin (mTOR) activity, promoted TFEB nucleus translocation, increased cathepsin B expression, enhanced autophagic degradation, and alleviated hepatic steatosis. No significant changes in phosphorylation of ULK1 in the hepatocytes were observed. However, when siRNA was used to knock down SIRT3, the changes of those protein were partially reversed, and hepatic steatosis was further exacerbated. CONCLUSIONS Our findings highlight irisin as a potential therapeutic for hepatic steatosis by modulating autophagy and lipid metabolism, potentially providing a novel therapeutic target for the management of MASLD. Further research is needed to elucidate the underlying mechanisms and explore the potential clinical applications of this approach in the treatment of MASLD.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jia Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Anran Ma
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhihong Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yunzhi Ni
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Di Wu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yue Zhou
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Na Zhang
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201203, China
| | - Li Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongsheng Chang
- Key Laboratory of Immune Microenvironment and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201203, China
| |
Collapse
|
18
|
Dye JA, Stewart EJ, Schladweiler MC, Nguyen HH, Grindstaff RD, Padgett WT, Fisher AA, Miller CN. Maternal Exposure to Ozone During Implantation Promotes a Feminized Transcriptomic Profile in the Male Adolescent Liver. Endocrinology 2025; 166:bqaf018. [PMID: 39865885 DOI: 10.1210/endocr/bqaf018] [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: 11/27/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Maternal exposure to ozone during implantation results in reduced fetal weight gain in rats. Offspring from ozone-exposed dams demonstrate sexually dimorphic risks to high-fat diet feeding in adolescence. To better understand the adolescent hepatic metabolic landscape following fetal growth restriction, RNA sequencing was performed to characterize the effects of ozone-induced fetal growth restriction on male and female offspring. Pregnant Long-Evans rats were exposed to filtered air or 0.8 ppm ozone for 4 hours on both gestation days 5 and 6 (n = 6/group). At approximately postnatal day 48, liver tissue was obtained for RNA sequencing from offspring. Peri-implantation exposure to ozone in the dam had greater effects on hepatic gene expression in male offspring than in the females. Interestingly, heatmaps of these differentially expressed genes suggested that male offspring from ozone-exposed dams had a transcriptomic pattern like that of female offspring. Using a filtered set of highly female-predominant genes (n = 390), 57% were upregulated in the male offspring from ozone-exposed dams. Upregulated canonical pathways included sirtuin and orexin signaling, estrogen receptor signaling, and integration of energy metabolism. Relatively few genes altered in the male offspring from ozone-exposed dams were associated with endpoints of sexual maturity, signifying the likely source of the observed feminization was not attributed to sex hormones. This study provides initial evidence that growth restriction in utero may increase the risk of hepatic feminization in male offspring. Additional work is needed to further understand the relationship between developmental undernutrition and feminization in the male liver.
Collapse
Affiliation(s)
- Janice A Dye
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Erica J Stewart
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Mette C Schladweiler
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Helen H Nguyen
- Oak Ridge Institute for Science and Education, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Rachel D Grindstaff
- Neuroendocrine Toxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - William T Padgett
- Neuroendocrine Toxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Anna A Fisher
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| | - Colette N Miller
- Cardiopulmonary Immunotoxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC 27711, USA
| |
Collapse
|
19
|
Yu L, Bao S, Zhu F, Xu Y, Zu F, Liu Y, Jiang R, Chen W, Chen S. Serum Lactate Dehydrogenase Is a Novel Predictor for the Severity in the Patients With MAFLD: A Cross-Sectional Study in Hefei, China. Diabetes Metab Syndr Obes 2025; 18:345-361. [PMID: 39931373 PMCID: PMC11807770 DOI: 10.2147/dmso.s492153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/28/2024] [Indexed: 02/13/2025] Open
Abstract
Background & Aims The aim of this study was to assess the association between the level of lactate dehydrogenase (LDH) and the severity of metabolic syndrome (MetS) and Metabolic Associated Fatty Liver Disease (MAFLD) and the potential diagnostic value of LDH for identifying at-risk metabolic associated steatohepatitis (MASH). Methods This cross-sectional, real-world retrospective study enrolled a total of 1118 obese patients in the department of bariatric surgery at the Second Affiliated Hospital of Anhui Medical University from January 1, 2018, to December 31, 2021. Of these, 855 were enrolled in the study cohort. MAFLD was defined as the presence or absence of fatty liver disease as suggested by histologic biopsy of liver or postoperative pathology slides, or even hematology, and meets one of the following three conditions: overweight or obesity, T2DM, and metabolic dysfunction (MetS). Serum LDH activity levels were measured in 885 patients, and logistic regression was used to analyze the relationship between LDH and metabolic syndrome and the severity of MAFLD. Results In the study cohort of 855 obese patients, 604 (70.6%) had MetS. Patients with MetS (214.1[209.0-219.2]) had significantly higher serum LDH levels than those without MetS (188.7[181.6-195.9]). Particularly, serum LDH level was significantly higher in subjects with hypertension, central obesity, diabetes mellitus or hyperglycemia, elevated levels of triglycerides, or reduced levels of high-density lipoprotein than in those without. Moreover, LDH concentrations were grouped according to the total number of MetS components present in each patient, with Serum LDH levels gradually increase with the total number of MetS components. The MASH subjects had significantly higher LDH levels than the other three less severe non-MASH cohorts, including normal liver, simple fatty steatosis, and B.MASH. Logistic regression showed that LDH was significantly and positively correlated with MAFLD, B.MASH, MASH, at-risk MASH, fibrosis grade ≥1, fibrosis grade ≥2 and fibrosis grade ≥3. Conclusion Increased LDH levels were significantly and independently associated with the presence and severity of metabolic syndrome and MAFLD, indicating that LDH could be used as a novel biomarker and clinical predictor of severity of metabolic syndrome and MAFLD.
Collapse
Affiliation(s)
- Liang Yu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Shiming Bao
- Department of Emergency Surgery, Tongling People’s Hospital, Tongling, People’s Republic of China
| | - Feng Zhu
- Department of General Surgery, Tongling People’s Hospital, Tongling, People’s Republic of China
| | - Yanyan Xu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Fuqiang Zu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Yanwei Liu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Runbeng Jiang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Wei Chen
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Song Chen
- Department of General Surgery, the Affiliated Chuzhou Hospital of Anhui Medical University (The First People’s Hospital of Chuzhou), Chuzhou, People’s Republic of China
| |
Collapse
|
20
|
Kineman RD, Del Rio-Moreno M, Waxman DJ. Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol 2025; 21:105-117. [PMID: 39322791 DOI: 10.1038/s41574-024-01037-0] [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] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; also known as nonalcoholic fatty liver disease) is a chronic condition associated with metabolic syndrome, a group of conditions that includes obesity, insulin resistance, hyperlipidaemia and cardiovascular disease. Primary growth hormone (GH) deficiency is associated with MASLD, and the decline in circulating levels of GH with weight gain might contribute to the development of MASLD. Raising endogenous GH secretion or administering GH replacement therapy in the context of MASLD enhances insulin-like growth factor 1 (IGF1) production and reduces steatosis and the severity of liver injury. GH and IGF1 indirectly control MASLD progression by regulating systemic metabolic function. Evidence supports the proposal that GH and IGF1 also have a direct role in regulating liver metabolism and health. This Review focuses on how GH acts on the hepatocyte in a sex-dependent manner to limit lipid accumulation, reduce stress, and promote survival and regeneration. In addition, we discuss how GH and IGF1 might regulate non-parenchymal cells of the liver to control inflammation and fibrosis, which have a major effect on hepatocyte survival and regeneration. Development of a better understanding of how GH and IGF1 coordinate the functions of specific, individual liver cell types might provide insight into the aetiology of MASLD initiation and progression and suggest novel approaches for the treatment of MASLD.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA.
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
21
|
Portela FS, Malheiro LFL, Oliveira CA, Mercês ÉAB, De Benedictis LM, De Benedictis JM, Fernandes AJV, Silva BS, Ávila JS, Correia TML, Oliveira MV, Silva Oliveira PD, Magalhães ACMD, Soares TDJ, Melo FFD, Amaral LSDB. High-intensity interval training improves hepatic redox status via Nrf2 downstream pathways and reduced CYP2E1 expression in female rats with cisplatin-induced hepatotoxicity. Food Chem Toxicol 2025; 196:115234. [PMID: 39746600 DOI: 10.1016/j.fct.2024.115234] [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: 11/19/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Cisplatin (CP) is an antineoplastic drug associated with various cytotoxic adverse effects, including hepatotoxicity. Exercise training may offer hepatoprotection by improving redox status. This study compared the effects of light-intensity continuous training (LICT), moderate-intensity continuous training (MICT), and high-intensity interval training (HIIT) on CP-induced hepatotoxicity in female Wistar rats. The rats were divided into five groups (n = 7): sedentary control (C + S), CP and sedentary (CP + S), CP with LICT (CP + LICT), CP with MICT (CP + MICT), and CP with HIIT (CP + HIIT). The training protocols involved eight weeks of treadmill exercise before CP administration (5 mg/kg). Seven days after CP injection, the rats were euthanized to collect blood and liver tissue samples. Our findings demonstrate that HIIT was the most effective protocol in preventing histopathological alterations and reducing oxidative and nitrosative damage markers in macromolecules, including 4-HNE (lipids), nitrotyrosine (proteins), and 8-OHdG (DNA). The reduction in these markers appears to be linked to decreased CYP2E1 levels. Moreover, HIIT activated the Nrf2 pathway and upregulated its downstream antioxidant enzymes, including SOD1, catalase, GPx, and HO-1. In conclusion, HIIT emerged as the most effective protocol for mitigating hepatic damage, likely through CYP2E1 suppression and enhancement of antioxidant defenses via Nrf2 signaling pathway activation.
Collapse
Affiliation(s)
- Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas - PPGM-SBFis. Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas - PPGM-SBFis. Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Érika Azenathe Barros Mercês
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Lais Mafra De Benedictis
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas - PPGM-SBFis. Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Júlia Mafra De Benedictis
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Ana Jullie Veiga Fernandes
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Bruna Santos Silva
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Júlia Spínola Ávila
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Thiago Macêdo Lopes Correia
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Márcio Vasconcelos Oliveira
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Patrícia da Silva Oliveira
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Amélia Cristina Mendes de Magalhães
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas - PPGM-SBFis. Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas - PPGM-SBFis. Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde - Campus Anísio Teixeira, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação em Biociências, Vitória da Conquista, Bahia, 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas - PPGM-SBFis. Vitória da Conquista, Bahia, 45029-094, Brazil
| |
Collapse
|
22
|
Arivazhagan L, Delbare S, Wilson RA, Manigrasso MB, Zhou B, Ruiz HH, Mangar K, Higa R, Brown E, Li H, Garabedian MJ, Ramasamy R, Moore KJ, Fisher EA, Theise ND, Schmidt AM. Sex differences in murine MASH induced by a fructose-palmitate-cholesterol-enriched diet. JHEP Rep 2025; 7:101222. [PMID: 39911943 PMCID: PMC11795143 DOI: 10.1016/j.jhepr.2024.101222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 02/07/2025] Open
Abstract
Background & Aims Metabolic syndrome-associated steatotic liver disease (MASLD) and metabolic syndrome-associated steatohepatitis (MASH) have global prevalence rates exceeding 25% and 3-6%, respectively. The introduction of high-fructose corn syrup to the diet in the 1970s has been linked to metabolic and hepatic disturbances. Despite these associations, the potential for sex-dependent responses resulting from fructose-containing diets on MASLD/MASH has not been addressed. Methods Female and male C57BL/6J mice were fed a fructose-palmitate-cholesterol (FPC)-NASH diet vs. standard chow for 16 weeks (n = 40 mice). At sacrifice, plasma and liver were retrieved, the latter for single-nucleus RNA sequencing. Publicly available data sets of human male and female MASH liver were probed. Results The FPC-NASH diet-induced metabolic dysfunction in both female and male mice, with females exhibiting more severe hepatic steatosis (p = 0.0262), inflammation (p = 0.0206), and fibrosis (p <0.0001). Single-nucleus RNA sequencing revealed distinct sex-specific transcriptional profiles in hepatocytes and stellate cells responding to the FPC-NASH diet compared to the standard chow. In female mice, compared to males, pathways associated with lipid and metabolic processes in hepatocytes and cell-cell communication and adhesion in stellate cells were enriched. Metabolic flux analyses demonstrated reduced bile acid metabolism in female mice and human hepatocytes in FPC-NASH and MASH conditions, respectively, compared to their male counterparts. Conclusions Molecular profiling of hepatocytes and stellate cells in FPC-NASH diet-fed mice revealed significant sex differences mirrored in human MASH. The identification of intrinsic, within-sex, diet-dependent disparities underscores the critical need to include both male and female individuals in MAFLD/MASH studies and clinical trials. Impact and implications Despite the importance of metabolic dysfunction-associated steatohepatitis (MASH) in impairment of human health, the potential for and mechanisms of sex-dependent responses have yet to be well-studied, particularly with respect to the possible influence of high-fructose corn syrup additives to the diet, which has been linked to metabolic and hepatic disturbances. In a mouse model of fructose supplementation to a NASH diet, female mice displayed significantly higher MASH scores (steatosis, inflammation and fibrosis) compared to male mice. Single-nucleus RNA sequencing of livers revealed intrinsic, diet-dependent molecular disparities within sex, which were exaggerated when comparing female vs. male mice fed the fructose-containing NASH diet; many of these findings were recapitulated in human female vs. male patients with MASH. These results highlight potential mechanistic explanations and therapeutic targets for addressing sex differences and underscore the need to study both sexes in animal models and human MASH.
Collapse
Affiliation(s)
- Lakshmi Arivazhagan
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Sofie Delbare
- NYU Cardiovascular Research Center, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Robin A. Wilson
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Michaele B. Manigrasso
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Boyan Zhou
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Henry H. Ruiz
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Kaamashri Mangar
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Ryoko Higa
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Emily Brown
- NYU Cardiovascular Research Center, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Huilin Li
- Departments of Population Health (Biostatistics) and Environmental Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Michael J. Garabedian
- Department of Microbiology, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Kathryn J. Moore
- NYU Cardiovascular Research Center, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Edward A. Fisher
- NYU Cardiovascular Research Center, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Neil D. Theise
- Department of Pathology, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine Langone Health, New York, NY 10016, USA
| |
Collapse
|
23
|
Yang C, Chen S, Feng B, Lu Y, Wang Y, Liao W, Wu S, Wang L. Association between menopause, body composition, and nonalcoholic fatty liver disease: A prospective cohort in northern China. Maturitas 2025; 192:108148. [PMID: 39571273 DOI: 10.1016/j.maturitas.2024.108148] [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: 07/12/2024] [Revised: 09/27/2024] [Accepted: 11/04/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND The association between menopause, changes in body composition, and nonalcoholic fatty liver disease is not clear, and there is a lack of weight management strategies for perimenopausal women from the perspective of preventing nonalcoholic fatty liver disease. METHODS A total of 1316 postmenopausal and 3049 premenopausal women in the Kailuan cohort in China between 2006 and 2017 were enrolled and followed up till 2021. Cox regression models, including the causal mediation analyses, were used to estimate the association between menopause and nonalcoholic fatty liver disease and the potential mediation effect of changes in body composition. We also explored the impact of weight changes on the correlation between menopause and nonalcoholic fatty liver disease. RESULTS Women who experienced menopause had a higher risk of nonalcoholic fatty liver disease than premenopausal women (9-year cumulative incidence: 56.87 % vs. 48.80 %, adjusted hazard ratio = 1.219, 95 % confidence interval: 1.088-1.365). The nine-year cumulative incidence of nonalcoholic fatty liver disease was higher among overweight/obese postmenopausal women (67.24 % vs. 45.74 %, P < 0.001) and those with abdominal obesity (63.36 % vs. 49.69 %, P < 0.001); however, the hazard ratio of menopause for nonalcoholic fatty liver disease was more evident in women with a body mass index under 23.0 kg/m2 (hazard ratio = 1.434, 95 % confidence interval: 1.168-1.759) and those with normal waist circumference (hazard ratio = 1.362, 95 % confidence interval: 1.129-1.643), which could partially be explained by the visceral fat index (7.09 % and 7.35 % mediation, respectively). Weight loss of 3 % or more or reduction in waist circumference by 5 % or more was associated with a 31.1 % reduction (95 % confidence interval, 20.8 %-40.0 %) or a 14.2 % reduction (95 % confidence interval, 1.1 %-25.6 %) in the risk of nonalcoholic fatty liver disease among the premenopausal women. For postmenopausal women, weight gain of 3 % or more was associated with an increased risk of nonalcoholic fatty liver disease, especially in individuals with a body mass index under 23.0 kg/m2. CONCLUSION Menopause was associated with a higher risk of nonalcoholic fatty liver disease, partially by increasing visceral fat. Controlling weight in perimenopausal women may reduce the risk.
Collapse
Affiliation(s)
- Chenlu Yang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China.
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, 57 Xinhua East Rd, Tangshan, Hebei Province 063000, China
| | - Baoyu Feng
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Ying Lu
- Department of Cardiology, Kailuan General Hospital, 57 Xinhua East Rd, Tangshan, Hebei Province 063000, China
| | - Yanhong Wang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China.
| | - Wei Liao
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China.
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, 57 Xinhua East Rd, Tangshan, Hebei Province 063000, China.
| | - Li Wang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China; State Key Laboratory of Respiratory Health and Multimorbidity, 5 Dong Dan San Tiao, Beijing 100005, China.
| |
Collapse
|
24
|
Kuniholm MH, Murenzi G, Shumbusho F, Brazier E, Plaisy MK, Mensah E, Wandeler G, Riebensahm C, Chihota BV, Samala N, Diero L, Semeere AS, Chanyachukul T, Borse R, Nguyen DTH, Perazzo H, Lopez-Iniguez A, Castilho JL, Maruri F, Jaquet A. Association of cardiovascular disease risk with liver steatosis and fibrosis in people with HIV in low- and middle-income countries. AIDS 2025; 39:11-21. [PMID: 39264586 PMCID: PMC11624086 DOI: 10.1097/qad.0000000000004012] [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: 02/09/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE The aim of this study was to understand the relationship between cardiovascular disease (CVD) risk and liver steatosis and fibrosis among people with HIV (PLWH) at least 40 years of age on antiretroviral therapy (ART) in low and middle-income countries (LMIC). DESIGN We used cross-sectional behavioral and clinical data collected during study enrollment visits in 2020-2022 for the Sentinel Research Network of International epidemiology Databases to Evaluate AIDS (SRN of IeDEA). METHODS Ten-year CVD risk was calculated using 2019 WHO nonlaboratory and laboratory models. Transient elastography was used to assess liver disease. Presence of steatosis and significant fibrosis were defined by controlled attenuation parameter (CAP) at least 248 dB/m and liver stiffness measurement (LSM) at least 7.1 kPa, respectively. Participants with viral hepatitis, hazardous alcohol consumption, and unsuppressed HIV viral load were excluded from the analysis. Logistic regression was used to estimate odds ratios, adjusting for study site, CD4 + T cell count, stavudine and didanosine exposure, and in models stratified by sex and geographic region. RESULTS There were 1750 participants from nine LMIC. Median CVD risk was 3% for both nonlaboratory and laboratory-based models. Adjusted odds ratios (ORs) for steatosis and significant fibrosis associated with laboratory CVD risk (≥10 vs. <5%) were OR = 1.83 [95% confidence interval (95% CI) = 1.21-2.76; P = 0.004] and OR = 1.62 (95% CI = 0.85-3.07; P = 0.14), respectively. Associations of CVD risk with steatosis were stronger in men and among participants at study sites outside Africa. CONCLUSION Higher CVD risk was associated with steatosis but not with significant fibrosis in PWH in our LMIC cohort.
Collapse
Affiliation(s)
- Mark H Kuniholm
- Department of Epidemiology and Biostatistics, University at Albany, State University of New York, Rensselaer, New York, USA
| | - Gad Murenzi
- Research for Development (RD Rwanda)
- Rwanda Military Hospital, Kigali, Rwanda
| | - Fabienne Shumbusho
- Research for Development (RD Rwanda)
- Rwanda Military Hospital, Kigali, Rwanda
| | - Ellen Brazier
- Institute for Implementation Science in Population Health
- Graduate School of Public Health and Health Policy, City University of New York, New York, New York, USA
| | - Marie K Plaisy
- National Institute for Health and Medical Research (INSERM) UMR 1219, Research Institute for Sustainable Development (IRD) EMR 271, University of Bordeaux, Bordeaux Population Health Centre, Bordeaux, France
| | | | - Gilles Wandeler
- Department of Infectious Diseases, Inselspital, Bern University Hospital
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Carlotta Riebensahm
- Department of Infectious Diseases, Inselspital, Bern University Hospital
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Belinda V Chihota
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Niharika Samala
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University, Indianapolis, Indiana, United States of America
| | | | - Aggrey S Semeere
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Rohidas Borse
- B.J. Government Medical College & Sassoon General Hospitals, Pune, Maharashtra, India
| | - Dung T H Nguyen
- Department of Infectious Diseases, National Hospital for Tropical Diseases, Hanoi, Vietnam
| | - Hugo Perazzo
- Evandro Chagas National Institute of Infectious Diseases -Oswaldo Cruz Foundation (INI/FIOCRUZ), Rio de Janeiro, Brazil
| | - Alvaro Lopez-Iniguez
- Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, Mexico City, Mexico
| | - Jessica L Castilho
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Fernanda Maruri
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Antoine Jaquet
- National Institute for Health and Medical Research (INSERM) UMR 1219, Research Institute for Sustainable Development (IRD) EMR 271, University of Bordeaux, Bordeaux Population Health Centre, Bordeaux, France
| |
Collapse
|
25
|
Kwon Y, Gottmann P, Wang S, Tissink J, Motzler K, Sekar R, Albrecht W, Cadenas C, Hengstler JG, Schürmann A, Zeigerer A. Induction of steatosis in primary human hepatocytes recapitulates key pathophysiological aspects of metabolic dysfunction-associated steatotic liver disease. J Hepatol 2025; 82:18-27. [PMID: 38977136 DOI: 10.1016/j.jhep.2024.06.040] [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: 09/21/2023] [Revised: 06/15/2024] [Accepted: 06/29/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common cause of chronic liver disease. Owing to limited available treatment options, novel pre-clinical models for target selection and drug validation are warranted. We have established and extensively characterized a primary human steatotic hepatocyte in vitro model system that could guide the development of treatment strategies for MASLD. METHODS Cryopreserved primary human hepatocytes from five donors varying in sex and ethnicity were cultured with free fatty acids in a 3D collagen sandwich for 7 days and the development of MASLD was followed by assessing classical hepatocellular functions. As proof of concept, the effects of the drug firsocostat (GS-0976) on in vitro MASLD phenotypes were evaluated. RESULTS Incubation with free fatty acids induced steatosis, insulin resistance, mitochondrial dysfunction, inflammation, and alterations in prominent human gene signatures similar to patients with MASLD, indicating the recapitulation of human MASLD in this system. The application of firsocostat rescued clinically observed fatty liver disease pathologies, highlighting the ability of the in vitro system to test the efficacy and potentially characterize the mode of action of drug candidates. CONCLUSIONS Altogether, our human MASLD in vitro model system could guide the development and validation of novel targets and drugs for the treatment of MASLD. IMPACT AND IMPLICATIONS Due to low drug efficacy and high toxicity, clinical treatment options for metabolic dysfunction-associated steatotic liver disease (MASLD) are currently limited. To facilitate earlier stop-go decisions in drug development, we have established a primary human steatotic hepatocyte in vitro model. As the model recapitulates clinically relevant MASLD characteristics at high phenotypic resolution, it can serve as a pre-screening platform and guide target identification and validation in MASLD therapy.
Collapse
Affiliation(s)
- Yun Kwon
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Pascal Gottmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany; German Institute of Human Nutrition (DIfE), Department of Experimental Diabetology, Nuthetal, Germany
| | - Surui Wang
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Joel Tissink
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Karsten Motzler
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Revathi Sekar
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Department of Toxicology, Dortmund, Germany
| | - Cristina Cadenas
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Department of Toxicology, Dortmund, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Department of Toxicology, Dortmund, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), Neuherberg, Germany; German Institute of Human Nutrition (DIfE), Department of Experimental Diabetology, Nuthetal, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
26
|
Pirola CJ, Landa MS, Schuman M, García SI, Salatino A, Sookoian S. Metabolic dysfunction-associated steatotic liver disease exhibits sex-specific microbial heterogeneity within intestinal compartments. Clin Mol Hepatol 2025; 31:179-195. [PMID: 39391907 PMCID: PMC11791572 DOI: 10.3350/cmh.2024.0359] [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: 05/15/2024] [Revised: 09/28/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND/AIMS Evidence suggests that the gastrointestinal microbiome plays a significant role in the biology of metabolic dysfunction-associated steatotic liver disease (MASLD). However, it remains unclear whether disparities in the gut microbiome across intestinal tissular compartments between the sexes lead to MASLD pathogenesis. METHODS Sex-specific analyses of microbiome composition in two anatomically distinct regions of the gut, the small intestine and colon, were performed using an experimental model of MASLD. The study involved male and female spontaneously hypertensive rats and the Wistar-Kyoto control rat strain, which were fed either a standard chow diet or a high-fat diet for 12 weeks to induce MASLD (12 rats per group). High-throughput 16S sequencing was used for microbiome analysis. RESULTS There were significant differences in the overall microbiome composition of male and female rats with MASLD, including variations in topographical gut regions. The beta diversity of the jejunal and colon microbiomes was higher in female rats than in male rats (PERMANOVA p-value=0.001). Sex-specific analysis and discriminant features using LEfSe showed considerable variation in bacterial abundance, along with distinct functional properties, in the jejunum and colon of animals with MASLD. Significantly elevated levels of lipopolysaccharide and protein expression of Toll-like receptor 4 were observed in the livers of male rats with MASLD compared with their female counterparts. CONCLUSION This study uncovered sexual dimorphism in the gut microbiome of MASLD and identified microbial heterogeneity within intestinal compartments. Insights into sex-specific variations in gut microbiome composition could facilitate customised treatment strategies.
Collapse
Affiliation(s)
- Carlos Jose Pirola
- Systems Biology of Complex Diseases, Translational Research in Health Center (CENITRES). Maimónides University, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Faculty of Health Science, Maimónides University, Buenos Aires, Argentina
| | - Maria Silvina Landa
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- University of Buenos Aires, School of Medicine, Institute of Medical Research A Lanari, Buenos Aires, Argentina
| | - Mariano Schuman
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- University of Buenos Aires, School of Medicine, Institute of Medical Research A Lanari, Buenos Aires, Argentina
| | - Silvia Inés García
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- University of Buenos Aires, School of Medicine, Institute of Medical Research A Lanari, Buenos Aires, Argentina
- Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina
| | - Adrian Salatino
- Max Planck Institute for Immunobiology and Epigenetics, Bioinformatics Facility, Frieburg, Germany
| | - Silvia Sookoian
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Faculty of Health Science, Maimónides University, Buenos Aires, Argentina
- Clinical and Molecular Hepatology, Translational Research in Health Center (CENITRES). Maimónides University, Buenos Aires, Argentina
| |
Collapse
|
27
|
Perez-Ternero C, Li W, Aubdool AA, Goldin RD, Loy J, Devalia K, Alazawi W, Hobbs AJ. Endogenous C-type natriuretic peptide offsets the pathogenesis of steatohepatitis, hepatic fibrosis, and portal hypertension. PNAS NEXUS 2025; 4:pgae579. [PMID: 39816244 PMCID: PMC11734523 DOI: 10.1093/pnasnexus/pgae579] [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: 08/30/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), hepatic fibrosis, and portal hypertension constitute an increasing public health problem due to the growing prevalence of obesity and diabetes. C-type natriuretic peptide (CNP) is an endogenous regulator of cardiovascular homeostasis, immune cell reactivity, and fibrotic disease. Thus, we investigated a role for CNP in the pathogenesis of MASLD. Wild-type (WT), global CNP (gbCNP-/-), and natriuretic peptide receptor-C (NPR-C-/-) knockout mice were fed a choline-deficient defined amino acid diet or administered CCl4. Liver damage was assessed by histological and biochemical analyses, with steatosis and portal vein size determined by ultrasound. Portal vein pressure and reactivity were measured in vivo and ex vivo, respectively. Pharmacological CNP delivery was used to evaluate prospective therapeutic benefit, and plasma CNP concentration was compared in controls and patients with cirrhosis. Circulating CNP concentration was lower in patients with cirrhosis compared with controls. gbCNP-/- mice were more susceptible, versus WT, to advanced steatohepatitis and hepatic fibrosis, characterized by increased immune cell infiltration, fibrosis, ballooning, plasma alanine aminotransferase concentration, and up-regulation of markers driving these processes. gbCNP-/- mice had increased portal vein diameter and pressure, underpinned by CNP insensitivity. NPR-C-/- animals recapitulated, comparatively, the exaggerated pathogenic phenotype in gbCNP-/- mice, whereas CNP reduced hepatic stellate cell proliferation via NPR-B-dependent inhibition of extracellular signal-related kinase 1/2. Administration of CNP reversed many aspects of disease severity. These data define a new intrinsic role for CNP in offsetting the pathogenesis of MASLD, hepatic fibrosis, and portal hypertension and the potential for targeting CNP signaling for treating these disorders.
Collapse
Affiliation(s)
- Cristina Perez-Ternero
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Aisah A Aubdool
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Robert D Goldin
- Centre for Pathology, St Mary’s Hospital, Imperial College, London W2 1NY, United Kingdom
| | - John Loy
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Adrian J Hobbs
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
28
|
Islam MA, Khairnar R, Fleishman J, Reznik SE, Ragolia L, Gobbooru S, Kumar S. Female C57BL/6 mice exhibit protection against nonalcoholic fatty liver disease and diabesity accompanied by differential regulation of hepatic lipocalin prostaglandin D 2 synthase. Mol Cell Endocrinol 2025; 595:112404. [PMID: 39505230 DOI: 10.1016/j.mce.2024.112404] [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: 09/22/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its development into nonalcoholic steatohepatitis (NASH) are challenging health concerns globally. Clinically, the prevalence and severity of NAFLD/NASH are higher in men than in premenopausal women. NAFLD is strongly correlated with obesity, both of which are tied to high-fat/fructose-rich western diets. Therefore, we aimed to investigate sexual dimorphism in NAFLD pathogenesis in male and female C57BL/6 mice fed different diets. Male and female C57BL/67 mice were divided into four groups and kept on a chow (C), chow plus high fructose (CF), high fat (HF), and high fat plus high fructose (HFF) diet for 22 weeks. Liver tissues were collected at the end of the study and processed for NAFLD/NASH-related histology (H&E and trichrome staining), protein expression (SREBP1, SCAP, FABP4, α-SMA, TGF-β and L-PGDS), and biochemical parameters measurement. Our results displayed that female mice exhibited protection against NAFLD and diabesity on HF and HFF diets compared to male mice fed similar diets. Additionally, female mice showed protection from fibrosis compared to male mice. Both male and female mice fed HF and HFF diet groups displayed the cytosol-to-nuclear translocation of Lipocalin Prostaglandin D2 Synthase (L-PGDS). Cytoplasmic levels of L-PGDS were absent in females compared to low levels in males, revealing a possible sex-specific mechanism tied to fructose and fat metabolism. Collectively, female mice showed protection against NAFLD and diabesity relative to male mice, accompanied by differential regulation of hepatic lipocalin prostaglandin D2 synthase.
Collapse
Affiliation(s)
- Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Joshua Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Louis Ragolia
- NYU Grossman Long Island School of Medicine, Mineola, NY, 11501, USA
| | - Shruthi Gobbooru
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
29
|
Yu H, Su X, Tao W, Sun W, Zhang X, Han Q, Zhao Z, Zhang Y, Chen X, Liu X, Jia D, Fang L, Li L. Prevalence and characteristics of liver steatosis and fibrosis in type 2 diabetes mellitus (T2DM) patients: a cross-sectional study in populations of eastern China. BMJ Open 2024; 14:e087550. [PMID: 39672583 PMCID: PMC11647396 DOI: 10.1136/bmjopen-2024-087550] [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: 04/12/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024] Open
Abstract
OBJECTIVES To describe the prevalence, clinical characteristics and risk factors of liver steatosis and fibrosis in type 2 diabetes mellitus (T2DM) patients in eastern China. DESIGN A cross-sectional, multicentre study based on an ongoing cohort study. SETTING 16 clinics in eastern China, including primary clinics to tertiary hospitals. PARTICIPANTS 1816 patients with T2DM diagnosis who met the inclusion criteria were recruited into the study. INTERVENTION Participants underwent elastography examination. MAIN OUTCOME MEASURES Descriptive analysis was performed to calculate the prevalence and characteristics of liver steatosis and fibrosis. The correlated factors were analysed using single- and multivariate logistic regression analysis. RESULTS The prevalence of liver steatosis in T2DM patients is 69.7%, with 46% moderate to severe steatosis. 34.6% and 6.7% of the patients were detected with liver fibrosis and cirrhosis. Steatosis patients were younger, had higher body mass index (BMI), higher levels of insulin resistance and more severe lipid metabolism disorders. Similar trends of differences were observed in patients with fibrosis. Female gender (OR=0.574, 95% CI 0.381 to 0.865), BMI (OR=1.491, 95% CI 1.375 to 1.616), disease duration, inflammation and serum lipid profile markers were risk factors of steatosis, while BMI (OR=1.204, 95% CI 1.137 to 1.275) and female gender (OR=0.672, 95% CI 0.470 to 0.961) were still the most significant predictors of liver fibrosis. CONCLUSIONS The prevalence of liver steatosis and fibrosis were high in patients with T2DM. Liver steatosis and fibrosis in these patients appeared to be more associated with lipid metabolism disorders and insulin resistance rather than glucose levels. TRIAL REGISTRATION NUMBER Clinical trial: NCT05597709.
Collapse
Affiliation(s)
- Hekai Yu
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
- Southeast University, Nanjing, Jiangsu, China
| | - Xianghui Su
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Xinjiang, China
| | - Wenxuan Tao
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
- Southeast University, Nanjing, Jiangsu, China
| | - Weixia Sun
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
- Southeast University, Nanjing, Jiangsu, China
| | - Xiaoyan Zhang
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
| | - Qing Han
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
| | - Zhuoxiao Zhao
- Nanjing Gaochun Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Yan Zhang
- Department of Endocrinology, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaoqian Chen
- Department of Endocrinology, Nanjing Central Hospital, Nanjing, Jiangsu, China
| | - Xinliang Liu
- Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Dianrong Jia
- Department of Endocrinology, Taizhou Jiangyan Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu, China
| | - Li Fang
- Nanjing Gaochun Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Ling Li
- Department of Endocrinology, School of Medicine, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
30
|
Ha S, Wong VWS, Zhang X, Yu J. Interplay between gut microbiome, host genetic and epigenetic modifications in MASLD and MASLD-related hepatocellular carcinoma. Gut 2024; 74:141-152. [PMID: 38950910 PMCID: PMC11671994 DOI: 10.1136/gutjnl-2024-332398] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/08/2024] [Indexed: 07/03/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a wide spectrum of liver injuries, ranging from hepatic steatosis, metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis to MASLD-associated hepatocellular carcinoma (MASLD-HCC). Recent studies have highlighted the bidirectional impacts between host genetics/epigenetics and the gut microbial community. Host genetics influence the composition of gut microbiome, while the gut microbiota and their derived metabolites can induce host epigenetic modifications to affect the development of MASLD. The exploration of the intricate relationship between the gut microbiome and the genetic/epigenetic makeup of the host is anticipated to yield promising avenues for therapeutic interventions targeting MASLD and its associated conditions. In this review, we summarise the effects of gut microbiome, host genetics and epigenetic alterations in MASLD and MASLD-HCC. We further discuss research findings demonstrating the bidirectional impacts between gut microbiome and host genetics/epigenetics, emphasising the significance of this interconnection in MASLD prevention and treatment.
Collapse
Affiliation(s)
- Suki Ha
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
31
|
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
|
32
|
Martínez‐Arenas L, Vinaixa C, Conde I, Lorente S, Díaz‐Fontenla F, Marques P, Pérez‐Rojas J, Montalvá E, Carvalho‐Gomes Â, Berenguer M. FibroScan compared to liver biopsy for accurately staging recurrent hepatic steatosis and fibrosis after transplantation for MASH. Liver Int 2024; 44:3174-3182. [PMID: 39225307 PMCID: PMC11586891 DOI: 10.1111/liv.16085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/03/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) recurrence after liver transplantation (LT) seems unavoidable and gradual. We aimed to evaluate the diagnostic accuracy in the post-LT setting of patients transplanted for metabolic dysfunction-associated steatohepatitis (MASH) of recurrent hepatic steatosis and fibrosis identified with FibroScan, compared to biopsy findings. METHODS This prospective cohort study included adults transplanted for MASH between 2010 and 2022 in three LT centres in Spain who underwent FibroScan and biopsy at least 1-year after LT. RESULTS In total, 44 patients transplanted for MASH after LT were included. The median time from LT to biopsy and FibroScan was 24.5 (interquartile range [IQR]:16-46) and 26.0 (IQR: 16.8-41.5) months, respectively. The median time between biopsy and FibroScan was 2.0 (IQR: 0-5) months. On FibroScan, significant steatosis was diagnosed in about half of the patients (n = 21, 47.7%), yet advanced fibrosis in only two cases (4.6%). On biopsy, a quarter of biopsied patients (n = 11, 25%) had a MASH diagnosis, two (4.6%) with significant fibrosis and one (2.3%) with cirrhosis. All patients with liver stiffness measurement (LSM) values <8 kPa (n = 35, 79.5%) had a fibrosis stage ≤F1 (negative predictive value = 100%). The combination of post-LT hypertension (odds ratio [OR]: 12.0, 95% confidence interval [CI]: 1.8-80.4, p = .010) and post-LT dyslipidaemia (OR: 7.9, 95% CI: 1.3-47.1, p = .024) with LSM (OR: 1.7, 95% CI: 1.1-2.8, p = .030) was independently associated with MASLD. CONCLUSIONS Although biopsy remains the gold standard for detecting fibrosis, our results suggest that LSM values <8 kPa after LT for MASH are strongly correlated with absence of significant/advanced fibrosis.
Collapse
Affiliation(s)
- Laura Martínez‐Arenas
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Department of BiotechnologyUniversitat Politècnica de ValènciaValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Vinaixa
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
- Hepatology and Liver Transplantation UnitHospital Universitario y Politécnico La FeValenciaSpain
| | - Isabel Conde
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
- Hepatology and Liver Transplantation UnitHospital Universitario y Politécnico La FeValenciaSpain
| | - Sara Lorente
- Hepatology and Liver Transplantation UnitHospital Clínico Universitario Lozano Blesa, Instituto de Investigación Sanitaria Aragón (IIS Aragón)ZaragozaSpain
| | - Fernando Díaz‐Fontenla
- Liver Unit and Digestive DepartmentHospital General Universitario Gregorio MarañónMadridSpain
| | - Patrice Marques
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
| | - Judith Pérez‐Rojas
- Department of PathologyHospital Universitario y Politécnico La FeValenciaSpain
| | - Eva Montalvá
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
- Hepatobiliopancreatic Surgery and Transplantation UnitHospital Universitario y Politécnico La FeValenciaSpain
- Department of SurgeryUniversitat de ValènciaValenciaSpain
| | - Ângela Carvalho‐Gomes
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
| | - Marina Berenguer
- Hepatology, Hepatobiliopancreatic Surgery and Transplant LaboratoryInstituto de Investigación Sanitaria La Fe (IIS La Fe)ValenciaSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Instituto de Salud Carlos IIIMadridSpain
- Hepatology and Liver Transplantation UnitHospital Universitario y Politécnico La FeValenciaSpain
- Department of MedicineUniversitat de ValènciaValenciaSpain
| |
Collapse
|
33
|
Colloca GA, Venturino A. Effect of Subgroups on Study Outcomes in Unresectable Hepatocellular Carcinoma Undergoing Upfront Systemic Treatment: A Meta-analysis. Am J Clin Oncol 2024; 47:585-590. [PMID: 38979979 DOI: 10.1097/coc.0000000000001133] [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: 07/10/2024]
Abstract
OBJECTIVES Immunotherapy improved the outcome of patients with unresectable hepatocellular carcinoma, but not all studies are in agreement, nor is it clear whether certain subgroups have really benefited. This study aims to perform an updated meta-analysis of trials comparing upfront immunotherapy-based regimens versus tyrosin-kinase inhibitors, and some exploratory analyses. METHODS After a systematic review, randomized trials of immunotherapy-based regimens versus tyrosin-kinase inhibitors were selected. A meta-analysis assessed the relationship between treatment arm and overall survival. Based on the resulting heterogeneity, a further investigation of 11 variables by meta-regression and an exploration of subgroups were planned. RESULTS Eight studies were selected. From the meta-analysis, the overall survival improvement for the immunotherapy-based arms was consistent (HR: 0.77, CI: 0.68-0.88), although heterogeneity between studies was significant ( Q =16.37; P =0.0373; I2 =51.1%). After meta-regression, the effect of the experimental arm was more pronounced in the elderly and lost among patients with HCV-related liver disease. Subgroups suggested a favorable effect of immunotherapy in patients with HBV-related hepatocellular carcinoma, extrahepatic dissemination, and elevated alpha-fetoprotein. CONCLUSION The study results confirm the significant overall survival improvement after immunotherapy-based regimens but suggest different effects on the outcome depending on age, etiology of liver disease, and tumor burden.
Collapse
|
34
|
Huang N, Su X, Yu T, Wu X, Lu B, Sun W, Yao L, Wang M, Wang Y, Wu W, Liu Y, Yang T, Gao R, Miao C, Li L. Serum 25-hydroxy vitamin D level is associated with elastography-detected liver fibrosis in patients with type 2 diabetes mellitus in China. Front Endocrinol (Lausanne) 2024; 15:1420088. [PMID: 39698035 PMCID: PMC11653015 DOI: 10.3389/fendo.2024.1420088] [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: 04/19/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024] Open
Abstract
Objective In this cross-sectional study including patients with type 2 diabetes mellitus (T2DM) we aimed to explore the relationship between serum 25-hydroxy vitamin (25(OH)D) level and liver steatosis and fibrosis in the Chinese population. Methods Patients visiting 16 clinical centers with T2DM were recruited. Their liver steatosis and fibrosis status were then assessed using elastography. Factors associated with steatosis and fibrosis were explored using regression analysis. Correlations between serum 25(OH)D levels and other patient characteristics were analyzed using linear regression. Results In total, 1,513 patients with T2DM were included in the study. The prevalence of steatosis and fibrosis was 69.7%, and 34.6%, separately. A lower level of 25(OH)D was detected in patients with liver steatosis compared to those without, although it was not an independent predictor of this condition. However, 25(OH)D level was independently associated with liver fibrosis even when adjusted for age, sex, body mass index, hemoglobin A1c, insulin, and homeostatic model assessment of insulin resistance (OR = 0.964 [0.935-0.993], P = 0.015). When patients were separated into subgroups by sex, a correlation between 25(OH)D and fibrosis was identified in the male group (OR = 0.969 [0.940-0.998], P = 0.038). Conclusions In conclusion, this multi-center, cross-sectional study in patients with T2DM showed that serum 25-hydroxy vitamin D level was strongly associated with liver fibrosis and this relationship was more pronounced in male patients. Clinical trial registration https://clinicaltrials.gov/ct2/show/, identifier NCT05597709.
Collapse
Affiliation(s)
- Nan Huang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Xianghui Su
- Department of Endocrinology, Changji Branch, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Ting Yu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaodong Wu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Bing Lu
- Department of Endocrinology, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Liqin Yao
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Maoyun Wang
- Department of Endocrinology, Baoying Hospital of Traditional Chinese Medicine, Yangzhou, China
| | - Yao Wang
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Wenxuan Wu
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Yingzhao Liu
- Department of Endocrinology, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Ting Yang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ruidong Gao
- Department of Endocrinology, Baoying Hospital of Traditional Chinese Medicine, Yangzhou, China
| | - Congqing Miao
- Department of Endocrinology, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| |
Collapse
|
35
|
Malheiro LFL, Oliveira CA, Portela FS, Mercês ÉAB, Benedictis LMD, Benedictis JMD, Andrade END, Magalhães ACM, Melo FFD, Oliveira PDS, Soares TDJ, Amaral LSDB. High-intensity interval training alleviates liver inflammation by regulating the TLR4/NF-κB signaling pathway and M1/M2 macrophage balance in female rats with cisplatin hepatotoxicity. Biochem Biophys Res Commun 2024; 733:150712. [PMID: 39317112 DOI: 10.1016/j.bbrc.2024.150712] [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: 07/31/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Cisplatin (CDDP) is an antineoplastic drug whose adverse effects include hepatotoxicity. The inflammatory process is crucial in the progression of liver injuries. Exercise is known for its anti-inflammatory effects, but the influence of different training modalities on hepatoprotection is still unclear. This study aims to compare the impacts between preconditioning with high-intensity interval training (HIIT) and traditional continuous training of low (LT) and moderate (MT) intensities on inflammatory markers in Wistar female rats with CDDP-induced hepatotoxicity. Thirty-five rats were divided into five groups: control and sedentary (C + Sed), treated with CDDP and sedentary (CDDP + Sed), treated with CDDP and subjected to LT (CDDP + LT), treated with CDDP and subjected to MT (CDDP + MT), and treated with CDDP and subjected to HIIT (CDDP + HIIT). The training protocols consisted of treadmill running for 8 weeks before CDDP treatment. The rats were euthanized 7 days after the treatment. Liver samples were collected to evaluate the expression of various inflammatory markers and types of macrophages. Our results indicated that HIIT was the only protocol to prevent the increase in all analyzed pro-inflammatory cytokines and reduce the number of ED-1-positive cells, attenuating the TLR4/NF-κB signaling pathway in the liver. Additionally, HIIT increased the anti-inflammatory cytokine IL-10 and regulated M1/M2 macrophage polarization. Thus, this study suggests that preconditioning with HIIT is more effective in promoting hepatoprotective effects than LT and MT, regulating inflammatory markers through modulation of the TLR4/NF-κB signaling pathway and M2 macrophage polarization in the hepatic tissue of female rats treated with CDDP.
Collapse
Affiliation(s)
- Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Fernanda Santos Portela
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Érika Azenatte Barros Mercês
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Laís Mafra de Benedictis
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Júlia Mafra de Benedictis
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | | | | | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Patrícia da Silva Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia, 45029-094, Brazil
| | | |
Collapse
|
36
|
Grayson C, Chalifoux O, Russo MDST, Avizonis DZ, Sterman S, Faerman B, Koufos O, Agellon LB, Mailloux RJ. Ablating the glutaredoxin-2 (Glrx2) gene protects male mice against non-alcoholic fatty liver disease (NAFLD) by limiting oxidative distress. Free Radic Biol Med 2024; 224:660-677. [PMID: 39278573 DOI: 10.1016/j.freeradbiomed.2024.09.016] [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: 07/23/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
In the present study, we investigated the consequences of deleting the glutaredoxin-2 gene (Glrx2-/-) on the development of non-alcoholic fatty liver disease (NAFLD) in male and female C57BL6N mice fed a control (CD) or high-fat diet (HFD). We report that the HFD induced a significant increase in body mass in the wild-type (Wt) and Glrx2-/- male, but not female, mice, which was associated with the hypertrophying of the abdominal fat. Interestingly, while the Wt male mice fed the HFD developed NAFLD, the deletion of the Glrx2 gene mitigated vesicle formation, intrahepatic lipid accumulation, and fibrosis in the males. The protective effect associated with ablating the Glrx2 gene in male mice was due to enhancement of mitochondrial redox buffering capacity. Specifically, liver mitochondria from male Glrx2-/- fed a CD or HFD produced significantly less hydrogen peroxide (mtH2O2), had lower malondialdehyde levels, greater activities for glutathione peroxidase and thioredoxin reductase, and less protein glutathione mixed disulfides (PSSG) when compared to the Wt male mice fed the HFD. These effects correlated with the S-glutathionylation of α-ketoglutarate dehydrogenase (KGDH), a potent mtH2O2 source and key redox sensor in hepatic mitochondria. In comparison to the male mice, both Wt and Glrx2-/- female mice displayed almost complete resistance to HFD-induced body mass increases and the development of NAFLD, which was attributed to the superior redox buffering capacity of the liver mitochondria. Together, our findings show that modulation of mitochondrial S-glutathionylation signaling through Glrx2 augments resistance of male mice towards the development of NAFLD through preservation of mitochondrial redox buffering capacity. Additionally, our findings demonstrate the sex dimorphisms associated with the manifestation of NAFLD is related to the superior redox buffering capacity and modulation of the S-glutathionylome in hepatic mitochondria from female mice.
Collapse
Affiliation(s)
- Cathryn Grayson
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Olivia Chalifoux
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Mariana De Sa Tavares Russo
- Goodman Cancer Institute, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada; Department of Medicine, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada
| | - Daina Zofija Avizonis
- Goodman Cancer Institute, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada; Department of Medicine, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada
| | - Samantha Sterman
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Ben Faerman
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Olivia Koufos
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Luis B Agellon
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada.
| |
Collapse
|
37
|
Kablawi D, Milic J, Thomas T, Fotsing Tadjo T, Cinque F, Elgretli W, Gioè C, Lebouché B, Tsochatzis E, Finkel J, Bhagani S, Cascio A, Guaraldi G, Mazzola G, Saeed S, Sebastiani G. Metabolic dysfunction-associated steatohepatitis exhibits sex differences in people with HIV. HIV Med 2024; 25:1259-1269. [PMID: 39091015 DOI: 10.1111/hiv.13697] [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: 06/06/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVES People with HIV are at increased risk for metabolic dysfunction-associated steatohepatitis (MASH). Although sex differences are documented in the general population, their role in the context of HIV is less understood. METHODS This was a multicentre cohort study including people with HIV without viral hepatitis coinfection. A FibroScan-AST (FAST) score >0.35 was used to diagnose MASH with significant liver fibrosis (stage F2-F4). We investigated sex-based differences in MASH trends as a function of age using a segmented linear mixed-effects model. Random effects accounted for clustering by the four sites. Adjusted models included ethnicity, diabetes, hypertension, and detectable HIV viral load. RESULTS We included 1472 people with HIV (25% women). At baseline, the prevalence of MASH with fibrosis by FAST score was lower in women than in men (4.8% vs. 9.2%, p = 0.008). Based on the adjusted model, male sex (+0.034; p = 0.04), age per year (+0.003; p = 0.05), detectable HIV viral load (+0.034; p = 0.02), and hypertension (+0.03; p = 0.01) were positively associated with MASH with fibrosis. Although men exhibited generally higher FAST scores, FAST scores increased in women during the critical biological age of presumed perimenopause to menopause (between 40 and 50 years), reaching levels similar to those in men by the age of 55 years. CONCLUSION Despite women with HIV having a lower prevalence of MASH with fibrosis than men, they exhibit an acceleration in FAST score increase around the perimenopausal age. Future studies should target adequate consideration of sex differences in clinical investigation of metabolic dysfunction-associated steatotic liver disease to fill current gaps and implement precision medicine for people with HIV.
Collapse
Affiliation(s)
- Dana Kablawi
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jovana Milic
- Modena HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena, Italy
| | - Tyler Thomas
- Public Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Thierry Fotsing Tadjo
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Felice Cinque
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Wesal Elgretli
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Claudia Gioè
- Infectious and Tropical Diseases Unit, Sicilian Regional Reference Center for the Fight against AIDS, AOU Policlinico "P. Giaccone", Palermo, Italy
| | - Bertrand Lebouché
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University Health Centre, Montreal, Quebec, Canada
- Center for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | - Antonio Cascio
- Infectious and Tropical Diseases Unit, Sicilian Regional Reference Center for the Fight against AIDS, AOU Policlinico "P. Giaccone", Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", Palermo, Italy
| | - Giovanni Guaraldi
- Modena HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Mazzola
- Department of Infectious Diseases, Sant'Elia Hospital, Caltanissetta, Italy
| | - Sahar Saeed
- Public Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Giada Sebastiani
- Chronic Viral Illness Service, Division of Infectious Disease, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Center for Outcomes Research and Evaluation, Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
38
|
Tai T, Shao YY, Zheng YQ, Jiang LP, Han HR, Yin N, Li HD, Ji JZ, Mi QY, Yang L, Feng L, Duan FY, Xie HG. Clopidogrel ameliorates high-fat diet-induced hepatic steatosis in mice through activation of the AMPK signaling pathway and beyond. Front Pharmacol 2024; 15:1496639. [PMID: 39508046 PMCID: PMC11537861 DOI: 10.3389/fphar.2024.1496639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Metabolic dysfunction-associated steatotic liver disease (MASLD) frequently confers an increased risk of vascular thrombosis; however, the marketed antiplatelet drugs are investigated for the prevention and treatment of MASLD in patients with these coexisting diseases. Methods To determine whether clopidogrel could ameliorate high-fat diet (HFD)-induced hepatic steatosis in mice and how it works, mice were fed on normal diet or HFD alone or in combination with or without clopidogrel for 14 weeks, and primary mouse hepatocytes were treated with palmitate/oleate alone or in combination with the compounds examined for 24 h. Body weight, liver weight, insulin resistance, triglyceride and total cholesterol content in serum and liver, histological morphology, transcriptomic analysis of mouse liver, and multiple key MASLD-associated genes and proteins were measured, respectively. Results and discussion Clopidogrel mitigated HFD-induced hepatic steatosis (as measured with oil red O staining and triglyceride kit assay) and reduced elevations in serum aminotransferases, liver weight, and the ratio of liver to body weight. Clopidogrel downregulated the expression of multiple critical lipogenic (Acaca/Acacb, Fasn, Scd1, Elovl6, Mogat1, Pparg, Cd36, and Fabp4), profibrotic (Col1a1, Col1a2, Col3a1, Col4a1, Acta2, and Mmp2), and proinflammatory (Ccl2, Cxcl2, Cxcl10, Il1a, Tlr4, and Nlrp3) genes, and enhanced phosphorylation of AMPK and ACC. However, compound C (an AMPK inhibitor) reversed enhanced phosphorylation of AMPK and ACC in clopidogrel-treated primary mouse hepatocytes and alleviated accumulation of intracellular lipids. We concluded that clopidogrel may prevent and/or reverse HFD-induced hepatic steatosis in mice, suggesting that clopidogrel could be repurposed to fight fatty liver in patients.
Collapse
Affiliation(s)
- Ting Tai
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Shao
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Yu-Qi Zheng
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacology, Nanjing Medical University School of Pharmacy, Nanjing, China
| | - Li-Ping Jiang
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Hao-Ru Han
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Na Yin
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Hao-Dong Li
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacology, Nanjing Medical University School of Pharmacy, Nanjing, China
| | - Jin-Zi Ji
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qiong-Yu Mi
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Li Yang
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Lei Feng
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Fu-Yang Duan
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
| | - Hong-Guang Xie
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China
- Department of Clinical Pharmacology, China Pharmaceutical University School of Basic Medicine and Clinical Pharmacy, Nanjing, China
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacology, Nanjing Medical University School of Pharmacy, Nanjing, China
| |
Collapse
|
39
|
Papadimitriou K, Mousiolis AC, Mintziori G, Tarenidou C, Polyzos SA, Goulis DG. Hypogonadism and nonalcoholic fatty liver disease. Endocrine 2024; 86:28-47. [PMID: 38771482 DOI: 10.1007/s12020-024-03878-1] [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: 01/15/2024] [Accepted: 05/12/2024] [Indexed: 05/22/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently proposed to be renamed to metabolic dysfunction-associated steatotic liver disease (MASLD), is a major global public health concern, affecting approximately 25-30% of the adult population and possibly leading to cirrhosis, hepatocellular carcinoma, and liver transplantation. The liver is involved in the actions of sex steroids via their hepatic metabolism and production of the sex hormone-binding globulin (SHBG). Liver disease, including NAFLD, is associated with reproductive dysfunction in men and women, and the prevalence of NAFLD in patients with hypogonadism is considerable. A wide spectrum of possible pathophysiological mechanisms linking NAFLD and male/female hypogonadism has been investigated. As therapies targeting NAFLD may impact hypogonadism in men and women, and vice versa, treatments of the latter may affect NAFLD, and an insight into their pathophysiological pathways is imperative. This paper aims to elucidate the complex association between NAFLD and hypogonadism in men and women and discuss the therapeutic options and their impact on both conditions.
Collapse
Affiliation(s)
- Kasiani Papadimitriou
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Athanasios C Mousiolis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gesthimani Mintziori
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
40
|
Ábel T, Benczúr B, Csobod ÉC. Sex differences in pathogenesis and treatment of dyslipidemia in patients with type 2 diabetes and steatotic liver disease. Front Med (Lausanne) 2024; 11:1458025. [PMID: 39376658 PMCID: PMC11456427 DOI: 10.3389/fmed.2024.1458025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
Previously published studies have shown that women with type 2 diabetes have a higher risk of atherosclerotic cardiovascular disease than men with type 2 diabetes. The exact reason for this is not yet known. The association between metabolic dysfunction-associated steatotic liver disease and type 2 diabetes appears to be bidirectional, meaning that the onset of one may increase the risk of the onset and progression of the other. Dyslipidemia is common in both diseases. Our aim was therefore to investigate whether there is a sex difference in the pathogenesis and management of dyslipidemia in patients with type 2 diabetes and steatotic liver disease with metabolic dysfunction. While the majority of published studies to date have found no difference between men and women in statin treatment, some studies have shown reduced effectiveness in women compared to men. Statin treatment is under-prescribed for both type 2 diabetics and patients with dysfunction-associated steatotic liver disease. No sex differences were found for ezetimibe treatment. However, to the best of our knowledge, no such study was found for fibrate treatment. Conflicting results on the efficacy of newer cholesterol-lowering PCSK9 inhibitors have been reported in women and men. Results from two real-world studies suggest that up-titration of statin dose improves the efficacy of PCSK9 inhibitors in women. Bempedoic acid treatment has been shown to be effective and safe in patients with type 2 diabetes and more effective in lipid lowering in women compared to men, based on phase 3 results published to date. Further research is needed to clarify whether the sex difference in dyslipidemia management shown in some studies plays a role in the risk of ASCVD in patients with type 2 diabetes and steatotic liver disease with metabolic dysfunction.
Collapse
Affiliation(s)
- Tatjana Ábel
- Department of Dietetics and Nutritional Sciences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Béla Benczúr
- Department of Dietetics and Nutritional Sciences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
- János Balassa County Hospital, Ist Department of Internal medicine (Cardiology/Nephrology), Szekszárd, Hungary
| | - Éva Csajbókné Csobod
- Department of Dietetics and Nutritional Sciences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
41
|
Yadav AK, MacNeill JJ, Krylov A, Ashrafi N, Mimi RA, Saxena R, Liu S, Graham SF, Wan J, Morral N. Sex- and age-associated factors drive the pathophysiology of MASLD. Hepatol Commun 2024; 8:e0523. [PMID: 39185904 PMCID: PMC11357696 DOI: 10.1097/hc9.0000000000000523] [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] [Received: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is strongly associated with obesity. Sex and age affect MASLD prevalence and pathophysiology. The use of animal models fed Western-style diets is vital for investigating the molecular mechanisms contributing to metabolic dysregulation and for facilitating novel drug target identification. However, the sex-associated and age-associated mechanisms underlying the pathophysiology remain poorly understood. This knowledge gap limits the development of personalized sex-specific and age-specific drug treatments. METHODS Young (7 wk) and aged (52 wk) male and female mice were fed a high-fat diet (HFD) or low-fat diet. Liver metabolome (>600 molecules) and transcriptome profiles were analyzed. RESULTS Male and female mice fed an HFD developed obesity, glucose intolerance, and hepatic steatosis. However, fasting blood glucose, insulin, and serum alanine aminotransferase levels were higher in males fed an HFD, indicating a more severe metabolic disease. In addition, males showed significant increases in liver diacylglycerides and glycosylceramides (known mediators of insulin resistance and fibrosis), and more changes in the transcriptome: extracellular matrix organization and proinflammatory genes were elevated only in males. In contrast, no major increase in damaging lipid classes was observed in females fed an HFD. However, aging affected the liver to a greater extent in females. Acylcarnitine levels were significantly reduced, suggestive of changes in fatty acid oxidation, and broad changes in the transcriptome were observed, including reduced oxidative stress response gene expression and alterations in lipid partitioning genes. CONCLUSIONS Here, we show distinct responses to an HFD between males and females. Our study underscores the need for using both sexes in drug target identification studies, and characterizing the molecular mechanisms contributing to the MASLD pathophysiology in aging animals.
Collapse
Affiliation(s)
- Ajay K. Yadav
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Justin J. MacNeill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aleksei Krylov
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nadia Ashrafi
- Metabolomics Department, Corewell Health Research Institute, Royal Oak, Michigan, USA
- Corewell Health William Beaumont University Hospital, Royal Oak, Michigan, USA
| | - Romana Ashrafi Mimi
- Metabolomics Department, Corewell Health Research Institute, Royal Oak, Michigan, USA
- Corewell Health William Beaumont University Hospital, Royal Oak, Michigan, USA
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stewart F. Graham
- Metabolomics Department, Corewell Health Research Institute, Royal Oak, Michigan, USA
- Corewell Health William Beaumont University Hospital, Royal Oak, Michigan, USA
- Oakland University-William Beaumont School of Medicine, Rochester, Michigan USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Núria Morral
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
42
|
Li X, He J, Sun Q. The prevalence and effects of sarcopenia in patients with metabolic dysfunction-associated steatotic liver disease (MASLD): A systematic review and meta-analysis. Clin Nutr 2024; 43:2005-2016. [PMID: 39053329 DOI: 10.1016/j.clnu.2024.07.006] [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: 04/28/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND AND AIMS Sarcopenia is a common complication in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). However, the prevalence and its impact on the survival of sarcopenia in patients with MASLD is unknown. In this study, we aimed to assess the prevalence and effects of sarcopenia in patients with MASLD. METHODS Systematic review and meta-analysis of full texts of relevant studies were searched from inception until June 12, 2024 in five databases (PubMed, Cochrane Library, Embase, Web of Science, and the China National Knowledge Infrastructure). Next, we assessed the prevalence of sarcopenia in MASLD, and calculated the ORs and HRs between sarcopenia and MASLD based on the adjusted data from individual studies. Statistical analyses were performed using Stata 11.0. RESULTS Of the 2984 records considered, 29 studies recruiting 63,330 patients were included. The pooled prevalence of sarcopenia in patients with MASLD was 23.5% overall (95% CI; 19.1%-27.9%, I2 = 99.6%), and was higher in Asian patients, male, cross-sectional studies, when BIA were employed to measure muscle mass, one criterion of diagnosis sarcopenia, MASLD was diagnosed employing MRI, and moderate-quality studies. Sarcopenia was associated with MASLD patients (adjusted odds ratio [aOR] 2.08, 95% CI 1.58-2.74, I2 = 93.6%) with similar findings in subgroups stratified by age, study design, methods for measuring muscle mass, assessment method to detect sarcopenia, and study quality. The association between all-cause mortality further supports the association between sarcopenia and poor prognosis with MASLD (aHR 1.59, 95% CI 1.33-1.91, I2 = 0%). CONCLUSIONS Sarcopenia was strongly associated with MASLD progression and was a risk factor not only for MASLD pathogenesis but was also markedly correlated with MASLD-associated mortality.
Collapse
Affiliation(s)
- Xiaoyan Li
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang, China
| | - Jie He
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, Sichuan, China; Clinical Medical College of Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Qiuhua Sun
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang, China.
| |
Collapse
|
43
|
Vitale A, Trapani S, Russo FP, Miele L, Svegliati Baroni G, Marchesini G, Burra P, Ottoveggio MS, Romagnoli R, Martini S, De Simone P, Carrai P, Cescon M, Morelli MC, De Carlis L, Belli L, Gruttadauria S, Volpes R, Colledan M, Fagiuoli S, Di Benedetto F, De Maria N, Rossi G, Caccamo L, Donato F, Vennarecci G, Di Costanzo GG, Vivarelli M, Carraro A, Sacerdoti D, Ettorre GM, Giannelli V, Agnes S, Gasbarrini A, Rossi M, Ginanni Corradini S, Mazzaferro V, Bhoori S, Manzia TM, Lenci I, Zamboni F, Mameli L, Baccarani U, Toniutto P, Lupo LG, Tandoi F, Rendina M, Andorno E, Giannini EG, Spada M, Billato I, Marchini A, Romano P, Brancaccio G, D’Amico F, Ricci A, Cardillo M, Cillo U, Associazione Italiana per lo Studio del Fegato (AISF)., Società Italiana Trapianti d’Organo (SITO)., Centro Nazionale Trapianti (CNT). Waiting list mortality and 5-year transplant survival benefit of patients with MASLD: An Italian liver transplant registry study. JHEP Rep 2024; 6:101147. [PMID: 39282226 PMCID: PMC11399673 DOI: 10.1016/j.jhepr.2024.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND & AIMS International consensus has recently introduced a new definition of metabolic dysfunction-associated steatotic liver disease (MASLD). We sought to analyse epidemiological trends, prognostic features, and transplant survival benefits of patients with MASLD and without MASLD waiting for liver transplantation (LT) in Italy. METHODS Using the Italian Liver Transplant Registry database, we analysed data from adult patients listed for primary LT attributable to end-stage chronic liver disease between January 2012 and December 2022. Independent multivariable waiting lists and post-transplant survival models were developed for patients with and without hepatocellular carcinoma (HCC). A Monte Carlo simulation was used to create 5-year transplant benefit distributions based on the presence of MASLD, HCC, and model for end-stage liver disease (MELD)-sodium values. RESULTS A total sample of 1,941 patients with MASLD and 11,201 patients without MASLD was considered. A significant increase in the prevalence of MASLD as an indication for LT was observed from 2012 to 2022, for both cohorts with HCC (from 17.7 to 30%) and without HCC (from 9.5 to 11.8%) cohorts. Projections suggest that, as early as next year, MASLD will overcome HCV as the second most common indication for transplantation after alcoholic liver disease in Italy. According to univariate and multivariate analyses, MASLD was not an independent predictive factor for patient survival after transplantation. However, it increased the risk of death for patients on the waiting list without HCC (hazard ratio 1.62, p <0.001). At the same MELD-sodium, the 5-year transplant benefit was higher in patients with non-HCC MASLD, followed by patients with HCC, whereas it was lower in patients without HCC and without MASLD. CONCLUSIONS Patients with non-HCC MASLD had an increased waitlist mortality and 5-year transplant survival benefit compared with other candidates. IMPACT AND IMPLICATIONS The present research addresses the critical need to understand the evolving landscape of liver transplantation indications, mainly focusing on metabolic dysfunction-associated steatotic liver disease (MASLD) in Italy. Given the significant rise in MASLD cases, these findings highlight that patients with non-HCC MASLD face increased waitlist mortality and benefit more from liver transplantation within 5 years compared with other candidates. The significance of these results lies in their emphasis on the necessity of focusing on patients with MASLD on waiting lists to improve outcomes. By tailoring transplant eligibility criteria and resource allocation, the study provides actionable insights to improve patient survival and optimise liver transplantation practices.
Collapse
Affiliation(s)
- Alessandro Vitale
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | - Silvia Trapani
- Centro Nazionale Trapianti, Istituto Superiore Di Sanitá, Italy
| | | | - Luca Miele
- Fondazione Policlinico Gemelli Irccs, Università Cattolica Del Sacro Cuore, Rome, Italy
| | | | - Giulio Marchesini
- Department of Medical and Surgical Sciences, Alma Mater University, IRCCS Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Patrizia Burra
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Salvatore Agnes
- Fondazione Policlinico Gemelli Irccs, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- Fondazione Policlinico Gemelli Irccs, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Massimo Rossi
- Sapienza Università Di Roma, Policlinico Umberto I, Rome, Italy
| | | | | | - Sherrie Bhoori
- Istituto Nazionale Tumori Milan and University of Milan, Milan, Italy
| | | | | | | | | | - Umberto Baccarani
- Dipartimento Di Area Medica, Università Degli Studi di Udine, Udine, Italy
| | - Pierluigi Toniutto
- Dipartimento Di Area Medica, Università Degli Studi di Udine, Udine, Italy
| | | | | | | | | | | | | | - Ilaria Billato
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | - Andrea Marchini
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | - Pierluigi Romano
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | | | - Francesco D’Amico
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | - Andrea Ricci
- Centro Nazionale Trapianti, Istituto Superiore Di Sanitá, Italy
| | | | - Umberto Cillo
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
| | - Associazione Italiana per lo Studio del Fegato (AISF).
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
- Centro Nazionale Trapianti, Istituto Superiore Di Sanitá, Italy
- Fondazione Policlinico Gemelli Irccs, Università Cattolica Del Sacro Cuore, Rome, Italy
- AOU Ospedali Riuniti, Ancona, Italy
- Department of Medical and Surgical Sciences, Alma Mater University, IRCCS Sant’Orsola-Malpighi Hospital, Bologna, Italy
- AOU Città della Salute, PO S.G.Battista, Torino, Italy
- AOU Pisana, Pisa, Italy
- AOU Sant’Orsola Malpighi, Bologna, Italy
- AO Niguarda Ca' Grada, Milano, Italy
- Is.Me.T.T., Palermo, Italy
- Ospedale Papa Giovanni XXIII, Bergamo, Italy
- Azienda Ospedaliera Policlinico, Modena, Italy
- Ospedale Maggiore Policlinico, Milan, Italy
- Azienda Ospedaliera ‘A. Cardarelli’, Italy
- Azienda Ospedaliera Verona, Verona, Italy
- Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
- Sapienza Università Di Roma, Policlinico Umberto I, Rome, Italy
- Istituto Nazionale Tumori Milan and University of Milan, Milan, Italy
- Azienda Ospedaliera Universitaria Policlinico Tor Vergata, Rome, Italy
- Azienda Ospedaliera G. Brotzu, Cagliari, Italy
- Dipartimento Di Area Medica, Università Degli Studi di Udine, Udine, Italy
- AOU Consorziale Policlinico di Bari, Bari, Italy
- A.O.U. S. Martino, Genova, Italy
- Ospedale Bambino Gesù, IRCCS, Rome, Italy
| | - Società Italiana Trapianti d’Organo (SITO).
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
- Centro Nazionale Trapianti, Istituto Superiore Di Sanitá, Italy
- Fondazione Policlinico Gemelli Irccs, Università Cattolica Del Sacro Cuore, Rome, Italy
- AOU Ospedali Riuniti, Ancona, Italy
- Department of Medical and Surgical Sciences, Alma Mater University, IRCCS Sant’Orsola-Malpighi Hospital, Bologna, Italy
- AOU Città della Salute, PO S.G.Battista, Torino, Italy
- AOU Pisana, Pisa, Italy
- AOU Sant’Orsola Malpighi, Bologna, Italy
- AO Niguarda Ca' Grada, Milano, Italy
- Is.Me.T.T., Palermo, Italy
- Ospedale Papa Giovanni XXIII, Bergamo, Italy
- Azienda Ospedaliera Policlinico, Modena, Italy
- Ospedale Maggiore Policlinico, Milan, Italy
- Azienda Ospedaliera ‘A. Cardarelli’, Italy
- Azienda Ospedaliera Verona, Verona, Italy
- Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
- Sapienza Università Di Roma, Policlinico Umberto I, Rome, Italy
- Istituto Nazionale Tumori Milan and University of Milan, Milan, Italy
- Azienda Ospedaliera Universitaria Policlinico Tor Vergata, Rome, Italy
- Azienda Ospedaliera G. Brotzu, Cagliari, Italy
- Dipartimento Di Area Medica, Università Degli Studi di Udine, Udine, Italy
- AOU Consorziale Policlinico di Bari, Bari, Italy
- A.O.U. S. Martino, Genova, Italy
- Ospedale Bambino Gesù, IRCCS, Rome, Italy
| | - Centro Nazionale Trapianti (CNT)
- Azienda Ospedale-Università Di Padova, Università di Padova, Padova, Italy
- Centro Nazionale Trapianti, Istituto Superiore Di Sanitá, Italy
- Fondazione Policlinico Gemelli Irccs, Università Cattolica Del Sacro Cuore, Rome, Italy
- AOU Ospedali Riuniti, Ancona, Italy
- Department of Medical and Surgical Sciences, Alma Mater University, IRCCS Sant’Orsola-Malpighi Hospital, Bologna, Italy
- AOU Città della Salute, PO S.G.Battista, Torino, Italy
- AOU Pisana, Pisa, Italy
- AOU Sant’Orsola Malpighi, Bologna, Italy
- AO Niguarda Ca' Grada, Milano, Italy
- Is.Me.T.T., Palermo, Italy
- Ospedale Papa Giovanni XXIII, Bergamo, Italy
- Azienda Ospedaliera Policlinico, Modena, Italy
- Ospedale Maggiore Policlinico, Milan, Italy
- Azienda Ospedaliera ‘A. Cardarelli’, Italy
- Azienda Ospedaliera Verona, Verona, Italy
- Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
- Sapienza Università Di Roma, Policlinico Umberto I, Rome, Italy
- Istituto Nazionale Tumori Milan and University of Milan, Milan, Italy
- Azienda Ospedaliera Universitaria Policlinico Tor Vergata, Rome, Italy
- Azienda Ospedaliera G. Brotzu, Cagliari, Italy
- Dipartimento Di Area Medica, Università Degli Studi di Udine, Udine, Italy
- AOU Consorziale Policlinico di Bari, Bari, Italy
- A.O.U. S. Martino, Genova, Italy
- Ospedale Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
44
|
Barr B, Gollahon L. The Modification of Dietary Protein with Ammonium Hydroxide Enhancement Improves Longevity and Metabolic Outcomes in a Sex-Dependent Manner. Nutrients 2024; 16:2787. [PMID: 39203925 PMCID: PMC11357104 DOI: 10.3390/nu16162787] [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: 07/26/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
(1) Background: Dietary protein is a key component of all dietary patterns. It has been demonstrated that there are subtle differences in health implications associated with the source of dietary protein consumed. This study examined dietary protein sources (DPSs) in a long-term study of diet-induced obesity ± ammonium hydroxide enhancement (AHE) and its role in improving long-term health outcomes. (2) Methods: Over 18 months, 272 C3H/HeJ mice (136 male and 136 female) were monitored on high-fat diets with varying DPSs ± AHE. Mice were monitored for weekly change in total mass, as well as 6-month assessments of lean and fat mass. At each assessment, a cohort (~8 mice per diet per sex) was censored for a cross-sectional examination of organ function. (3) Results: Longevity was improved in females fed AHE diets, regardless of DPSs. Females' measures of fat and lean mass were markedly elevated with casein protein diets compared to beef protein diets regardless of AHE. Females fed a beef protein diet + AHE demonstrated reduced fat mass and increased lean mass with aging. In males, AHE beef protein diet-fed mice showed marked improvement to longevity and increased lean mass at 6 months. (4) Conclusions: This study demonstrates that dietary protein modification by AHE attenuates the negative impacts of HF diets in both males and females in a sex-dependent manner. Furthermore, the results from this study emphasize the importance of identifying the differences in the utilization of dietary proteins in both a sex- and age-related manner and demonstrate the potential of DPS modification by AHE as a dietary intervention.
Collapse
Affiliation(s)
- Benjamin Barr
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| |
Collapse
|
45
|
Chen H, Zhou Y, Hao H, Xiong J. Emerging mechanisms of non-alcoholic steatohepatitis and novel drug therapies. Chin J Nat Med 2024; 22:724-745. [PMID: 39197963 DOI: 10.1016/s1875-5364(24)60690-4] [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: 07/02/2024] [Indexed: 09/01/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a leading cause of chronic liver disease globally. It initiates with simple steatosis (NAFL) and can progress to the more severe condition of non-alcoholic steatohepatitis (NASH). NASH often advances to end-stage liver diseases such as liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Notably, the transition from NASH to end-stage liver diseases is irreversible, and the precise mechanisms driving this progression are not yet fully understood. Consequently, there is a critical need for the development of effective therapies to arrest or reverse this progression. This review provides a comprehensive overview of the pathogenesis of NASH, examines the current therapeutic targets and pharmacological treatments, and offers insights for future drug discovery and development strategies for NASH therapy.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Zhou
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
46
|
Becker EC, Siddique O, O'Sullivan DM, Dar W, Einstein M, Morgan G, Emmanuel B, Sotil EU, Richardson E, Serrano OK. Disparities in Liver Transplantation for Nonalcoholic Steatohepatitis in Women. Transplantation 2024; 108:e181-e186. [PMID: 38419160 DOI: 10.1097/tp.0000000000004964] [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: 03/02/2024]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is the fastest-growing indication for liver transplantation (LT). Sex disparities among patients with cirrhosis on the LT waitlist are well known. We wanted to understand these disparities further in women with end-stage liver disease patients listed for NASH cirrhosis in a contemporary cohort. METHODS We used data from the Scientific Registry of Transplant Recipients to assess sex racial, and ethnic differences in NASH patients listed for LT. Adults transplanted from August 1997 to June 2021 were included. Inferential statistics were used to evaluate differences with univariate and multivariate comparisons, including competitive risk analysis. RESULTS During the study time period, we evaluated 12 844 LT for NASH cirrhosis. Women were transplanted at a lower rate (46.5% versus 53.5%; P < 0.001) and higher model for end-stage liver disease (MELD) (23.8 versus 22.6; P < 0.001) than men. Non-White women were transplanted at a higher MELD (26.1 versus 23.1; P < 0.001) than White women and non-White male patients (26.1 versus 24.8; P < 0.001). Graft and patient survivals were significantly different ( P < 0.001) between non-White women and White women and men (White and non-White). CONCLUSIONS Evaluation of LT candidates in the United States demonstrates women with NASH cirrhosis have a higher MELD than men at LT. Additional disparities exist among non-White women with NASH as they have higher MELD and creatinine at LT compared with White women. After LT, non-White women have worse graft and patient survival compared with men or White women. These data indicate that non-White women with NASH are the most vulnerable on the LT waitlist.
Collapse
Affiliation(s)
- Erica C Becker
- Department of Internal Medicine, University of Connecticut Health, Farmington, CT
| | - Osama Siddique
- Department of Gastroenterology, Hartford Hospital, Hartford, CT
| | - David M O'Sullivan
- Department of Research Administration, Hartford HealthCare, Hartford, CT
| | - Wasim Dar
- Transplant Program, Hartford Hospital, Hartford, CT
- Department of Surgery, University of Connecticut Health, Farmington, CT
| | - Michael Einstein
- Department of Internal Medicine, University of Connecticut Health, Farmington, CT
- Transplant Program, Hartford Hospital, Hartford, CT
| | - Glyn Morgan
- Transplant Program, Hartford Hospital, Hartford, CT
- Department of Surgery, University of Connecticut Health, Farmington, CT
| | - Bishoy Emmanuel
- Transplant Program, Hartford Hospital, Hartford, CT
- Department of Surgery, University of Connecticut Health, Farmington, CT
| | - Eva U Sotil
- Department of Internal Medicine, University of Connecticut Health, Farmington, CT
- Transplant Program, Hartford Hospital, Hartford, CT
| | - Elizabeth Richardson
- Department of Internal Medicine, University of Connecticut Health, Farmington, CT
- Transplant Program, Hartford Hospital, Hartford, CT
| | - Oscar K Serrano
- Transplant Program, Hartford Hospital, Hartford, CT
- Department of Surgery, University of Connecticut Health, Farmington, CT
| |
Collapse
|
47
|
Guaraldi G, Milic J, Renzetti S, Motta F, Cinque F, Bischoff J, Desilani A, Conti J, Medioli F, del Monte M, Kablawi D, Elgretli W, Calza S, Mussini C, Rockstroh JK, Sebastiani G. The effect of weight gain and metabolic dysfunction-associated steatotic liver disease on liver fibrosis progression and regression in people with HIV. AIDS 2024; 38:1323-1332. [PMID: 38597416 PMCID: PMC11216384 DOI: 10.1097/qad.0000000000003903] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 04/11/2024]
Abstract
OBJECTIVE People with HIV (PWH) have high risk of liver fibrosis. We investigated the effect of weight gain and metabolic dysfunction-associated steatotic liver disease (MASLD) on liver fibrosis dynamics. DESIGN Multicenter cohort study. METHODS Fibrosis progression was defined as development of significant fibrosis [liver stiffness measurement (LSM) ≥8 kPa], or transition to cirrhosis (LSM ≥13 kPa), for those with significant fibrosis at baseline. Fibrosis regression was defined as transition to LSM less than 8 kPa, or to LSM less than 13 kPa for those with cirrhosis at baseline. MASLD was defined as hepatic steatosis (controlled attenuation parameter >248 dB/m) with at least one metabolic abnormality. A continuous-time multistate Markov model was used to describe transitions across fibrosis states. RESULTS Among 1183 PWH included from three centers (25.2% with viral hepatitis coinfection), baseline prevalence of significant fibrosis and MASLD was 14.4 and 46.8%, respectively. During a median follow-up of 2.5 years (interquartile range 1.9-3.5), the incidence rate of fibrosis progression and regression was 2.8 [95% confidence interval (CI) 2.3-3.4] and 2.2 (95% CI 1.9-2.6) per 100 person-years, respectively. In Markov model, weight gain increased the odds of fibrosis progression [odds ratio (OR) 3.11, 95% CI 1.59-6.08], whereas weight gain (OR 0.30, 95% CI 0.10-0.84) and male sex (OR 0.32, 95% CI 0.14-0.75) decreased the odds of fibrosis regression. On multivariable Cox regression analysis, predictors of fibrosis progression were weight gain [adjusted hazard ratio (aHR) 3.12, 95% CI 1.41-6.90] and MASLD (aHR 2.72, 95% CI 1.05-7.02). CONCLUSION Fibrosis transitions are driven by metabolic health variables in PWH, independently of viral hepatitis coinfection and antiretroviral class therapy.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- Modena HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena
| | - Jovana Milic
- Modena HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena
| | - Stefano Renzetti
- Department of Medical-Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia
| | - Federico Motta
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Modena, Italy
| | - Felice Cinque
- Division of Gastroenterology and Hepatology, McGill University Health Centre
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, Canada
| | - Jenny Bischoff
- Department of Internal Medicine I, Venusberg Campus 1, University Hospital Bonn, Bonn Germany
| | - Andrea Desilani
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| | - Jacopo Conti
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| | - Filippo Medioli
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| | - Martina del Monte
- Infectious Diseases Clinic, Azienda Ospedaliero-Universitaria, Policlinico of Modena, Modena, Italy
| | - Dana Kablawi
- Division of Gastroenterology and Hepatology, McGill University Health Centre
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, Canada
| | - Wesal Elgretli
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Stefano Calza
- Unit of Biostatistics and Bioinformatics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Mussini
- Modena HIV Metabolic Clinic, University of Modena and Reggio Emilia, Modena
| | - Juergen K. Rockstroh
- Department of Internal Medicine I, Venusberg Campus 1, University Hospital Bonn, Bonn Germany
| | - Giada Sebastiani
- Division of Gastroenterology and Hepatology, McGill University Health Centre
- Chronic Viral Illness Service, Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
48
|
Bin DH, Liu F, Peng KP, Zhan M, Tan Y, Liu Q, Tang W, Mo ZN, Peng XJ, Tian GX. The relationship between follicle-stimulating hormone and metabolic dysfunction-associated fatty liver disease in men. Nutr Diabetes 2024; 14:52. [PMID: 38991999 PMCID: PMC11239811 DOI: 10.1038/s41387-024-00314-1] [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: 10/17/2023] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVES The present study aimed to investigate the relationship between male hormones and metabolic dysfunction-associated fatty liver disease (MAFLD) in males. METHODS Data from the Fangchenggang Area Male Health and Examination Survey (FAMHES) were used to analyze the male hormone levels between MAFLD patients and controls. Univariate and multivariate logistic regression analyses were performed to identify risk factors for MAFLD. Receiver operating characteristic curve analysis was used to assess the diagnostic performance of male hormones for MAFLD. RESULT A total of 1578 individuals were included, with 482 individuals (30.54%) of MAFLD, including 293 (18.57%) with mild disease and 189 (11.98%) with moderate-to-severe disease. The MAFLD patients were significantly older than those without MAFLD. The LH, FSH, and SHBG levels in the MAFLD patients were significantly greater than those in the control group. Age, FSH, LH, SHBG, and estradiol were all risk factors for MAFLD. Age, FSH, and LH were risk factors for moderate-to-severe MAFLD. FSH was an independent risk factor for MAFLD and moderate-to-severe MAFLD. FSH showed an excellent diagnostic value, with an AUC of 0.992 alone and 0.996 after adjusting age. CONCLUSIONS Our findings indicate that FSH may be a potential diagnostic and predictive biomarker for MAFLD.
Collapse
Affiliation(s)
- Dong-Hua Bin
- Department of Anus and Intesine, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Fang Liu
- Department of Ultrasoud, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Ke-Ping Peng
- Department of Otorhinolaryngology-Head and Neck surgery, The first Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Min Zhan
- Department of Anus and Intesine, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yan Tan
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiao Liu
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wang Tang
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zeng-Nan Mo
- Centre for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Xiong-Jun Peng
- Department of Medical Equipment, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Gui-Xiang Tian
- Department of Ultrasoud, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
49
|
Rodríguez-Hernández N, Lazo-de-la-Vega-Monroy ML, Ruiz-Noa Y, Preciado-Puga MDC, Garcia-Ramirez JR, Jordan-Perez B, Garnelo-Cabañas S, Ibarra-Reynoso LDR. Predictive Models for Non-Alcoholic Fatty Liver Disease Diagnosis in Mexican Patients with Gallstone Disease: Sex-Specific Insights. Diagnostics (Basel) 2024; 14:1487. [PMID: 39061624 PMCID: PMC11275442 DOI: 10.3390/diagnostics14141487] [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: 05/14/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Evidence regarding Non-Alcoholic Fatty Liver Disease (NAFLD) diagnosis is limited in the context of patients with gallstone disease (GD). This study aimed to assess the predictive potential of conventional clinical and biochemical variables as combined models for diagnosing NAFLD in patients with GD. (2) Methods: A cross-sectional study including 239 patients with GD and NAFLD diagnosed by ultrasonography who underwent laparoscopic cholecystectomy and liver biopsy was conducted. Previous clinical indices were also determined. Predictive models for the presence of NAFLD stratified by biological sex were obtained through binary logistic regression and sensitivity analyses were performed. (3) Results: For women, the model included total cholesterol (TC), age and alanine aminotransferase (ALT) and showed an area under receiver operating characteristic curve (AUC) of 0.727 (p < 0.001), sensitivity of 0.831 and a specificity of 0.517. For men, the model included TC, body mass index (BMI) and aspartate aminotransferase (AST), had an AUC of 0.898 (p < 0.001), sensitivity of 0.917 and specificity of 0.818. In both sexes, the diagnostic performance of the designed equations was superior to the previous indices. (4) Conclusions: These models have the potential to offer valuable guidance to healthcare providers in clinical decision-making, enabling them to achieve optimal outcomes for each patient.
Collapse
Affiliation(s)
- Nemry Rodríguez-Hernández
- Department of Medical Sciences, Health Sciences Division, University of Guanajuato, Leon Campus, Leon de los Aldama 37320, Mexico; (N.R.-H.); (M.-L.L.-d.-l.-V.-M.); (Y.R.-N.)
| | - María-Luisa Lazo-de-la-Vega-Monroy
- Department of Medical Sciences, Health Sciences Division, University of Guanajuato, Leon Campus, Leon de los Aldama 37320, Mexico; (N.R.-H.); (M.-L.L.-d.-l.-V.-M.); (Y.R.-N.)
| | - Yeniley Ruiz-Noa
- Department of Medical Sciences, Health Sciences Division, University of Guanajuato, Leon Campus, Leon de los Aldama 37320, Mexico; (N.R.-H.); (M.-L.L.-d.-l.-V.-M.); (Y.R.-N.)
| | - Monica-del-Carmen Preciado-Puga
- Department of Medicine and Nutrition, Health Sciences Division, University of Guanajuato, Leon Campus, Leon de los Aldama 37320, Mexico; (M.-d.-C.P.-P.); (J.-R.G.-R.)
| | - Juana-Rosalba Garcia-Ramirez
- Department of Medicine and Nutrition, Health Sciences Division, University of Guanajuato, Leon Campus, Leon de los Aldama 37320, Mexico; (M.-d.-C.P.-P.); (J.-R.G.-R.)
| | - Benjamin Jordan-Perez
- Department of Surgery, General Hospital Leon, Leon de los Aldama 37320, Mexico; (B.J.-P.); (S.G.-C.)
| | - Serafin Garnelo-Cabañas
- Department of Surgery, General Hospital Leon, Leon de los Aldama 37320, Mexico; (B.J.-P.); (S.G.-C.)
| | - Lorena-del-Rocío Ibarra-Reynoso
- Department of Medical Sciences, Health Sciences Division, University of Guanajuato, Leon Campus, Leon de los Aldama 37320, Mexico; (N.R.-H.); (M.-L.L.-d.-l.-V.-M.); (Y.R.-N.)
| |
Collapse
|
50
|
Booijink R, Ramachandran P, Bansal R. Implications of innate immune sexual dimorphism for MASLD pathogenesis and treatment. Trends Pharmacol Sci 2024; 45:614-627. [PMID: 38853100 DOI: 10.1016/j.tips.2024.05.004] [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: 04/10/2024] [Revised: 04/18/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Growing evidence suggests that metabolic dysfunction-associated steatotic liver disease (MASLD) is significantly higher in men versus women. Increased prevalence is observed in postmenopausal women, suggesting that age and sex (hormones) influence MASLD development and progression. Molecular data further reveal that sex regulates the innate immune responses with an essential role in MASLD progression. To date, there has been limited focus on the role of innate immune sexual dimorphism in MASLD, and differences between men and women are not considered in the current drug discovery landscape. In this review, we summarize the sex disparities and innate immune sexual dimorphism in MASLD pathogenesis. We further highlight the importance of harnessing sexual dimorphism in identifying therapeutic targets, developing pharmacological therapies, and designing (pre-) clinical studies for the personalized treatment for MASLD.
Collapse
Affiliation(s)
- Richell Booijink
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Prakash Ramachandran
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, UK
| | - Ruchi Bansal
- Personalized Diagnostics and Therapeutics, Department of Bioengineering Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|