1
|
Yang CY, Chen JH, Chen CY, Kao CY, Huang SF, Chang WY, Tu HP, Huang JF, Yu ML, Tai CM. Serial changes in metabolic dysfunction-associated steatotic liver disease after sleeve gastrectomy and their associations with abdominal adiposity: a prospective cohort study. Surg Obes Relat Dis 2025; 21:537-546. [PMID: 39706718 DOI: 10.1016/j.soard.2024.11.007] [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: 07/09/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Little is known about the associations between changes in hepatic steatosis and changes in abdominal adiposity after metabolic bariatric surgery. OBJECTIVES To evaluate the serial changes in hepatic steatosis and abdominal adiposity following sleeve gastrectomy (SG). SETTING University hospital, Taiwan. METHODS In this prospective study, patients who underwent SG and intraoperative liver biopsy were enrolled. Magnetic resonance imaging (MRI) was performed to assess the liver fat fraction (LFF), visceral adipose tissue (VAT) area, and subcutaneous adipose tissue (SAT) area. Liver fibrosis was assessed preoperatively via biopsy and the fibrosis-4 index (FIB-4) and postoperatively with the FIB-4. RESULTS Seventy-six metabolic dysfunction-associated steatotic liver disease (MASLD) patients, including 67 pure MASLD patients and 9 MASLD patients with combined etiologies, were enrolled. LFF and visceral-to-subcutaneous fat ratio were associated with metabolic dysfunction-associated steatohepatitis, and VAT area was associated with significant fibrosis (≥F2). Twelve months after SG, all MRI measurements significantly improved. The median LFF of pure MASLD patients decreased from 17.4% at baseline to 4.2% and 3.7% at the 6th and 12th postoperative months, respectively. Complete resolution of steatosis was achieved in 97.5% of patients at the 12th postoperative months. Using %VAT and %SAT reductions at the sixth postoperative month as references, LFF decreased more rapidly, with fold ratios of 1.3 and 1.8, respectively. CONCLUSIONS SG resulted in a significant decrease in hepatic steatosis and abdominal adiposity in patients with severe obesity, but hepatic steatosis improved faster than abdominal adiposity. Hepatic steatosis resolved in almost all patients 12 months after SG.
Collapse
Affiliation(s)
- Chung-Yi Yang
- Department of Medical imaging, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan
| | - Jian-Han Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; Division of General Surgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Chung-Yen Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; Division of General Surgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Cheng-Yi Kao
- Department of Medical imaging, E-DA Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Shiu-Feng Huang
- Investigator and Attending Physician, Institute of Molecular and Genomic Medicine, National Health Research Institutes
| | - Wen-Yu Chang
- Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; Department of Dermatology, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Pin Tu
- Department of Public Health and Environmental Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center of Hepatitis Research, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center of Hepatitis Research, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chi-Ming Tai
- Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.
| |
Collapse
|
2
|
Souza M, Al-Sharif L, Diaz I, Mantovani A, Villela-Nogueira CA. Global Epidemiology and Implications of PNPLA3 I148M Variant in Metabolic Dysfunction-Associated Steatotic Liver Disease: A Systematic Review and Meta-analysis. J Clin Exp Hepatol 2025; 15:102495. [PMID: 39882540 PMCID: PMC11773032 DOI: 10.1016/j.jceh.2024.102495] [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: 08/03/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025] Open
Abstract
Background & Aims PNPLA3 rs738409 variant is a risk factor for onset and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). We aimed to assess its global prevalence, clinical and histological characteristics, and long-term outcomes in patients with MASLD. Methods PubMed and Embase databases were searched until December 30, 2023, for observational studies on PNPLA3 genotyped adults with MASLD. Proportions were pooled using a generalized linear mixed model with Clopper-Pearson intervals. Continuous and dichotomous variables were analyzed using the DerSimonian-Laird method. International Prospective Register of Systematic Reviews registration number: CRD42023449838. Results A total of 109 studies involving 118,302 individuals with MASLD were identified. The overall minor allele frequency of the G allele [MAF(G)] at PNPLA3 was 0.45 (95% confidence interval [CI]: 0.43; 0.48) with high heterogeneity (I2 = 98%). The highest MAF(G) was found in Latin America (0.63) and the lowest in Europe (0.38). No African countries were identified. Carriers of the PNPLA3 variant had reduced adiposity, altered fat metabolism, and worse liver damage/histology than noncarriers. There was significant heterogeneity in the clinical/histological analyses (I2 > 50%). Only the PNPLA3 GG genotype was associated with higher mortality and liver-related events with no heterogeneity (I2 = 0%). Metaregressions showed the influence of adiposity, age, diabetes mellitus, and glucose on some PNPLA3 expression parameters. Overall, there was a moderate risk of bias in the included studies. Conclusions This study reveals the global pattern of PNPLA3 and its clinical, histological, and outcome implications in MASLD. Patients with MASLD and PNPLA3 variant have different clinical features and worse liver severity, and only PNPLA3 GG has a higher risk of mortality and liver outcomes. Our findings highlight the importance of PNPLA3 genotyping in clinical trials and advocate for personalized medicine approaches.
Collapse
Affiliation(s)
- Matheus Souza
- Department of Internal Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ivanna Diaz
- Department of Internal Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, United States
| | - Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | | |
Collapse
|
3
|
Macêdo APA, de Sousa Neto IV, Antonio GCF, Gaspar RC, de Lima RD, Dias LM, Vieira RFL, Muñoz VR, Brunelli DT, da Silva ASR, Cintra DE, Ropelle ER, Pauli JR. Time-restricted feeding reduces inflammatory markers and downregulates JAG1 and NICD protein levels in the liver of aged mice. Nutrition 2025; 133:112691. [PMID: 39983606 DOI: 10.1016/j.nut.2025.112691] [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: 10/14/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 02/23/2025]
Abstract
OBJECTIVES The present study aimed to assess whether Time-Restricted Feeding (TRF) modulates inflammation and hepatic Notch1 signalling in C57BL/6J-aged mice. METHODS Adult mice submitted to the ad libitum diet, aged (24 months-old) submitted to the ad libitum diet and, aged-TRF (24 months-old) subjected to the TRF (12 hours fed in the active cycle and 12 hours fasting in the light cycle) for 8 weeks. We investigated metabolic parameters, liver histology, metabolic-dysfunction-associated fatty liver disease activity score, collagen fiber, hepatic mitochondrial respiration, and publicly available liver Rna-seq datasets from human livers in diverse clinical conditions to clarify Notch1 involvement in liver health. RESULTS Our results demonstrated that aged mice (24 months old) showed increases in body weight, liver mass, Notch1 intracellular domain (NICD), and inflammatory markers (NFκB and TLR4 protein levels) in the liver when compared to adult animals. On the other hand, aged mice submitted to a TRF protocol showed reductions in inflammation and collagen fibers, which was accompanied by lower protein content of JAGGED1 and NICD in the liver. Furthermore, aged-TRF mice demonstrated increased liver mitochondrial respiration coupled with ATP production compared to the aged groups. Publicly available liver RNA-seq datasets in humans support our findings, indicating the upregulation of NOTCH1 in fibrosis and inflammation development. CONCLUSIONS TRF can reduce inflammatory markers and protein content of JAGGED1 and NICD in the liver of aged mice, which can contribute to tissue health and cellular longevity.
Collapse
Affiliation(s)
- Ana Paula Azevêdo Macêdo
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto (EEFERP), University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Rafael Calais Gaspar
- Departments of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robson Damasceno de Lima
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Larissa Moreira Dias
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- School of Physical Education and Sport of Ribeirão Preto (EEFERP), University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Diego Trevisan Brunelli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto (EEFERP), University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics (LabGeN), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
4
|
Gao Z, Cao S, Yuan H, Wu JZ, Zou G. Broad antifibrotic activities of AK3280 in pulmonary, hepatic, cardiac, and skin fibrosis animal models. Int Immunopharmacol 2025; 151:114337. [PMID: 40015207 DOI: 10.1016/j.intimp.2025.114337] [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: 12/12/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 03/01/2025]
Abstract
Fibrosis is the pathological outcome of many chronic inflammatory diseases, affecting various human organs. It is a significant contributor to global morbidity and mortality that affects nearly half of the elderly population. Pirfenidone (PFD) and nintedanib are approved by the FDA for treating pulmonary fibrosis, but these treatments are associated with poor tolerability and limited efficacy. Moreover, no antifibrotic drugs are approved for other fibrosis-related diseases, highlighting an urgent unmet medical need for more effective therapies. Here we report the in vivo pharmacological activities of AK3280, a novel, orally bioavailable small molecule designed to enhance pharmacokinetics, antifibrotic activity, and tolerability over PFD. AK3280 demonstrated antifibrotic effects across multiple organs, including the lungs, liver, heart, and skin, in various animal models. These results suggest that AK3280 holds promise as a clinically beneficial antifibrotic therapy for a range of fibrotic diseases, especially pulmonary, hepatic, cardiac, and skin fibrosis.
Collapse
Affiliation(s)
- Zhao Gao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Sushan Cao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Haiqing Yuan
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Jim Zhen Wu
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China
| | - Gang Zou
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai 201203, China.
| |
Collapse
|
5
|
Li JZ, Yang L, Xiao MX, Li N, Huang X, Ye LH, Zhang HC, Liu ZQ, Li JQ, Liu YY, Liang XJ, Li TY, Li JY, Cao Y, Pan Y, Lin XG, Dai HM, Dai EH, Li MR. Spatial and Single-Cell Transcriptomics Reveals the Regional Division of the Spatial Structure of MASH Fibrosis. Liver Int 2025; 45:e16125. [PMID: 39400982 DOI: 10.1111/liv.16125] [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: 02/18/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To elucidate the regional distribution of metabolic dysfunction-associated steatohepatitis (MASH) fibrosis within the liver and to identify potential therapeutic targets for MASH fibrosis. METHODS Liver sections from healthy controls, patients with simple steatosis and MASH patients were analysed using spatial transcriptomics integrated with single-cell RNA-seq. RESULTS Spatial transcriptomics analysis of liver tissues revealed that the fibrotic region (Cluster 9) was primarily distributed in lobules, with some fibrosis also found in the surrounding area. Integration of the single-cell-sequencing data set (GSE189175) showed a greater proportion of inflammatory cells (Kupffer cells and T cells) and myofibroblasts in MASH. Six genes, showing high- or low-specific expression in Cluster 9, namely, ADAMTSL2, PTGDS, S100A6, PPP1R1A, ASS1 and G6PC, were identified in combination with pathology. The average expression levels of ADAMTSL2, PTGDS and S100A6 on the pathological HE staining map were positively correlated with the increase in the degree of fibrosis and aligned strongly with the distribution of fibrosis. ADAMTSL2+ myofibroblasts play a role in TNF signalling pathways and in the production of ECM structural components. Pseudotime analysis indicated that in the early stages of MASH, infiltration by T cells and Kupffer cells triggers a significant inflammatory response. Subsequently, this inflammation leads to the activation of hepatic stellate cells (HSCs), transforming them into myofibroblasts and promoting the development of liver fibrosis. CONCLUSION This study is the first to characterise lineage-specific changes in gene expression, subpopulation composition, and pseudotime analysis in MASH fibrosis and reveals potential therapeutic targets for this condition.
Collapse
Affiliation(s)
- Jin-Zhong Li
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liu Yang
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Min-Xi Xiao
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ni Li
- Division of General Internal Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Xin Huang
- Division of Hepatobiliary Surgery, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Li-Hong Ye
- Division of Pathology, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Hai-Cong Zhang
- Division of Pathology, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Zhi-Quan Liu
- Division of Pathology, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Jun-Qing Li
- Division of Liver Disease, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Yun-Yan Liu
- Division of Liver Disease, The Fifth Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, China
| | - Xu-Jing Liang
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tao-Yuan Li
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jie-Ying Li
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yang Cao
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yun Pan
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xun-Ge Lin
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hai-Mei Dai
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Er-Hei Dai
- Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, China
| | - Min-Ran Li
- Division of Infectious Disease, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
6
|
Hu W, Wang H, Gan L, Lin Y, Fu Y, Tan W, Dou X, Ye L. Sulfonation of Capsaicin by sulfotransferases produces an anti-inflammatory metabolite with NF-κB pathway modulatory activity. Fitoterapia 2025; 182:106463. [PMID: 40090597 DOI: 10.1016/j.fitote.2025.106463] [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: 12/17/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025]
Abstract
Capsaicin (CAP), the principal bioactive component of chili peppers (Capsicum annuum L.), is widely recognized for its anti-inflammatory properties. However, its oral bioavailability is low, likely due to extensive sulfonation metabolism. Despite the well-known pharmacological benefits of CAP, the role of sulfotransferase (SULT)-mediated sulfonation in modulating its therapeutic effects remains poorly understood. This study aims to elucidate the sulfonate metabolic profile of CAP, investigate the anti-inflammatory role of its sulfonate metabolite (CAP-S), and uncover the mechanisms underlying CAP-S's anti-inflammatory effects. In our study, the mono-sulfonate metabolite of CAP, designated as CAP-S ((E)-N-[(4-sulfo-3-methoxyphenyl)methyl]-8-methylnon-6-enamide), is identified using ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) and proton nuclear magnetic resonance (1H-NMR). The metabolic profile of CAP was investigated in liver S9 fractions from human, rat, and mouse samples, with sulfonation of CAP examined using seven major recombinant SULT isoforms. The results demonstrate that CAP is primarily catalyzed by SULT1A subfamily and SULT1E1. The anti-inflammatory effects of CAP-S are evaluated in lipopolysaccharide (LPS)-stimulated RAW264.7 cells and an acute liver injury (ALI) mouse model. CAP-S significantly reduces inflammatory mediators and nitric oxide (NO) production in LPS-induced RAW264.7 cells. In vivo, CAP-S treatment alleviates hepatocyte necrosis, inflammatory cell infiltration, and reduces aspartate aminotransferase, alanine aminotransferase, and malondialdehyde levels, while enhancing superoxide dismutase activity and decreasing NO production. Additionally, CAP-S exerts comparable anti-inflammatory effects to CAP by suppressing NF-κB p65 phosphorylation and reducing pro-inflammatory cytokines, as evidenced by network pharmacology and western blot assays. These findings underscore the role of sulfonation in modulating CAP's therapeutic potential.
Collapse
Affiliation(s)
- Wanyu Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongyu Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lili Gan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yating Lin
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yufang Fu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weiling Tan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xianrui Dou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan 528300, China.
| | - Ling Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
7
|
Coombes JD, Manka PP, Swiderska-Syn M, Vannan DT, Riva A, Claridge LC, Moylan C, Suzuki A, Briones-Orta MA, Younis R, Kitamura N, Sydor S, Bittencourt S, Mi Z, Kuo PC, Diehl AM, van Grunsven LA, Chokshi S, Canbay A, Abdelmalek MF, Aspichueta P, Papa S, Eksteen B, Syn WK. Osteopontin Promotes Cholangiocyte Secretion of Chemokines to Support Macrophage Recruitment and Fibrosis in MASH. Liver Int 2025; 45:e16131. [PMID: 39422353 DOI: 10.1111/liv.16131] [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: 03/21/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND AND AIMS Osteopontin (OPN) promotes the ductular reaction and is a major driver of chronic liver disease (CLD) progression. Although CLD is characterised by the accumulation of inflammatory cells including macrophages around the peri-portal regions, the influence of OPN on recruitment is unclear. We investigated the role of OPN in cholangiocyte chemokine production and macrophage recruitment by combining in vivo, in vitro, and in silico approaches. METHODS The effects of OPN on cholangiocyte chemokine production and macrophage migration were assessed in culture, alongside RNA-sequencing to identify genes and pathways affected by OPN depletion. Murine liver injury models were used to assess liver chemokine expression and liver macrophage/monocyte recruitment. OPN and chemokine expression were analysed in liver tissue and plasma from biopsy-proven metabolic dysfunction-associated alcoholic steatohepatitis (MASH) patients. RESULTS OPN-knockdown in cholangiocytes reduced chemokine secretion. RNA-sequencing showed OPN-related effects clustered around immunity, chemotaxis and chemokine production. Macrophage exposure to cholangiocyte-conditioned media showed OPN-supported migration via chemokines chemokine (C-C motif) ligand (CCL)2, CCL5 and chemokine (C-X-C motif) ligand (CXCL)1. These effects were related to NF-κB signalling. Murine liver fibrosis was accompanied by upregulated liver OPN, CCL2, CCL5 and CXCL1 mRNA, and accumulation of liver cluster of differentiation (CD)11b/F4/80+CC chemokine receptors (CCR2)high macrophages but treatment with OPN-specific neutralising aptamers reduced fibrosis, chemokine mRNAs and accumulation of liver CD11b/F4/80+CCR2high/lymphocyte antigen 6 complexhigh inflammatory monocytes. In human MASH, liver OPN correlated with chemokines CCL2 and IL8 in association with portal injury and fibrosis. Plasma OPN, serum CCL2 and IL8 also increased with fibrosis stage. CONCLUSIONS OPN promotes cholangiocyte chemokine secretion and the accumulation of pro-inflammatory monocytes. These data support neutralisation of OPN as an anti-inflammatory and anti-fibrotic strategy.
Collapse
Affiliation(s)
- Jason D Coombes
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences and Medicine, King's College London, London, UK
- Division of Gastroenterology and Hepatology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Paul P Manka
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences and Medicine, King's College London, London, UK
- Gastroenterology and Hepatology, University Clinic Bochum, Bochum, Germany
| | - Marzena Swiderska-Syn
- Division of Gastroenterology and Hepatology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Danielle T Vannan
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Aspen Woods Clinic, Calgary, Alberta, Canada
| | - Antonio Riva
- Faculty of Life Sciences and Medicine, King's College London, London, UK
- Viral Hepatitis and Alcohol Research Group, Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Lee C Claridge
- Department of Hepatology, Leeds Teaching Hospital NHS Trust, Leeds, UK
| | - Cynthia Moylan
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Ayako Suzuki
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Marco A Briones-Orta
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Rasha Younis
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Naoto Kitamura
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Svenja Sydor
- Gastroenterology and Hepatology, University Clinic Bochum, Bochum, Germany
| | | | - Zhiyong Mi
- Department of Surgery, University of South Florida, Tampa, Florida, USA
| | - Paul C Kuo
- Department of Surgery, University of South Florida, Tampa, Florida, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina, USA
| | | | - Shilpa Chokshi
- Faculty of Life Sciences and Medicine, King's College London, London, UK
- Viral Hepatitis and Alcohol Research Group, Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Ali Canbay
- Gastroenterology and Hepatology, University Clinic Bochum, Bochum, Germany
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, EPV/EHU, Leioa, Spain
| | - Salvatore Papa
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Bertus Eksteen
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Aspen Woods Clinic, Calgary, Alberta, Canada
| | - Wing-Kin Syn
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, UK
- Division of Gastroenterology and Hepatology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, EPV/EHU, Leioa, Spain
| |
Collapse
|
8
|
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
|
9
|
Zhang J, Chen L, Zhao C, Chen Z, Xiao S, Yin X, Wu N, Yang L, Xu J, Zhou H, Wu Q, Shao R, Xu W. Polysaccharides from Cynanchum auriculatum Royle ex Wight ameliorate symptoms of hyperglycemia by regulating gut microbiota in type 2 diabetes mellitus mice. Int J Biol Macromol 2025; 299:139878. [PMID: 39818385 DOI: 10.1016/j.ijbiomac.2025.139878] [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: 01/06/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Type 2 diabetes mellitus (T2DM) represents a chronic metabolic disorder characterized by disrupted carbohydrate and lipid balance, resulting in hyperglycemia. This study evaluated the impact of polysaccharides derived from Cynanchum auriculatum Royle ex Wight (CRP) on mitigating hyperglycemia and modulating intestinal microbiota in T2DM mice. Findings indicated that CRP is mainly linked by →6)α-D-Glcp-(1→ and CRP-H demonstrated greater efficacy than CRP-L in regulating hypoglycemic-related indicators such as serum high-density lipoprotein cholesterol (HDL-c) level. Additionally, CRP at varying doses enhanced the mRNA expression of insulin receptor substrate 1 (IRS-1), phosphatidylinositol 3-kinase (PI3K), protein kinase B (AKT-1), and glucose transporter 2 (GLUT-2). Following a 4-week CRP-H treatment, a significant reduction in the Firmicutes/Bacteroidetes ratio at the phylum level was observed, alongside a marked increase in the relative abundance of beneficial genera such as Limosillactobacillus and Prevotella. Overall, CRP-H displayed enhanced hypoglycemic properties by activating the IRS-1/PI3K/AKT-1/GLUT-2 pathway and enriching beneficial gut bacteria, including Prevotella and Limosillactobacillus. This study establishes a foundational framework for further development and application of Cynanchum auriculatum Royle ex Wight resources, emphasizing the hypoglycemic potential of CRP.
Collapse
Affiliation(s)
- Jiawei Zhang
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Ligen Chen
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Chengyu Zhao
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Zhuo Chen
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Shiqi Xiao
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Xuemei Yin
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Na Wu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Lei Yang
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Jianda Xu
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, Changzhou 213003, China
| | - Hongcheng Zhou
- School of Medicine, Jiangsu Medical College, Yancheng 224051, China
| | - Qin Wu
- School of Medicine, Jiangsu Medical College, Yancheng 224051, China
| | - Rong Shao
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China
| | - Wei Xu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng 224051, China.
| |
Collapse
|
10
|
Loh Jiezhen T, Salomao M, Moreira R, Pai R. Reliability of an Expanded Non-alcoholic Steatohepatitis Grading and Staging System for Assessment of Disease Activity and Fibrosis. Int J Surg Pathol 2025; 33:281-288. [PMID: 39034304 DOI: 10.1177/10668969241260226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The nonalcoholic steatohepatitis clinical research network (NASH-CRN) system is commonly used for histologic assessment of disease activity and fibrosis in NASH. Despite this, the system does not fully capture the range of disease activity and fibrosis. As such, an expanded NAS (E-NAS) grading and staging system with a calculated E-NAS index was developed by our group. In this follow up study, we aim to revalidate the E-NAS system and compare its reliability to existing systems. Hematoxylin and eosin and trichrome stained sections from 40 liver biopsies were reviewed digitally by four hepatopathologists and assessed using the NASH-CRN and E-NAS systems as well as a modified Ishak fibrosis stage. The pathologist's gestalt impression of disease activity and fibrosis was scored on a Visual Analogue Scale (VAS), which ranged from 0 (no activity/fibrosis) to 100 (the worst activity/fibrosis ever seen). Inter-rater reliability was assessed, and Spearman correlation coefficients were calculated. The E-NAS index had higher inter-rater agreement versus the NAS score (ICC 0.70 vs 0.61). The inter-rater agreement for ballooning in the E-NAS system was also higher at 0.67 compared to the NAS (ICC 0.60). ICCs for fibrosis were comparable between all the systems assessed (0.78 to 0.88). Finally, the calculated E-NAS index was higher with increasing stage of fibrosis compared to the NAS suggesting that it associates better with fibrosis. In summary, the E-NAS system demonstrates substantial inter-rater reliability as well as improved correlation with disease activity VAS and fibrosis compared to the NAS score.
Collapse
Affiliation(s)
- Tracy Loh Jiezhen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Marcela Salomao
- Department of Pathology and Laboratory Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Roger Moreira
- Department of Pathology and Laboratory Medicine, Mayo Clinic Rochester, Rochester, MN, USA
| | - Rish Pai
- Department of Pathology and Laboratory Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| |
Collapse
|
11
|
Gaspar RC, Macêdo APA, Nakandakari SCBR, Muñoz VR, Abud GF, Vieira RFL, de Sousa Neto IV, Pavan ICB, da Silva LGS, Simabuco FM, da Silva ASR, Junior WS, Marchini JS, Nonino CB, Cintra DE, Ropelle ER, Pajvani UB, de Freitas EC, Pauli JR. Notch1 Signalling Is Downregulated by Aerobic Exercise, Leading to Improvement of Hepatic Metabolism in Obese Mice. Liver Int 2025; 45:e70068. [PMID: 40078075 DOI: 10.1111/liv.70068] [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: 11/01/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Notch1 protein plays a significant role in hepatic metabolism, as evidenced by its correlation with insulin resistance in the livers of obese individuals, making it an intriguing research target. Therefore, this study aims to investigate the impact of aerobic exercise on Notch1 pathways in the hepatic tissue of obese mice and its role in controlling hepatic metabolism. METHODS Therefore, we conducted a cross-sectional study utilising liver biopsies from lean and obese humans, as well as an intervention study involving mice subjected to a high-fat diet. The obese-trained mice group underwent a treadmill-running protocol for 4 weeks. RESULTS Our findings revealed that obese individuals exhibited increased NOTCH1 mRNA levels compared to lean subjects. The detrimental effects of Notch1 signalling were confirmed by Notch1-overexpressed HepG2 cell lines. Obese mice with higher hepatic Notch1 signalling demonstrated a reduction in this pathway when subjected to a 4-week treadmill running. Another benefit noticed in this trained group was the amelioration of insulin resistance, as well as a reduction in pyruvate intolerance and gluconeogenic enzymes. Additionally, we observed that these protective findings were accompanied by a decrease in mTORC1 pathway activity and lipid accumulation in the liver. Pharmacological inhibition of Notch1 in obese mice led to an increase in mitochondrial respiration in the liver. CONCLUSIONS We conclude that Notch1 signalling may be a potentially useful therapeutic target in obesity, while aerobic exercise training suppresses the Notch1 pathway in the liver, contributing to the regulation of hepatic glucose and lipid metabolism in obese mice.
Collapse
Affiliation(s)
- Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Ana Paula Azevêdo Macêdo
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | | | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Gabriela Ferreira Abud
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Applied Molecular Signaling Laboratory (LabSIMA), Department of Biochemistry, Federal University of São Paulo, Campinas, Sao Paulo, Brazil
| | - Adelino S R da Silva
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Wilson Salgado Junior
- School of Medicine of Ribeirão Preto, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Julio Sergio Marchini
- Department of Internal Medicine, Division of Nutrology, Ribeirao Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Carla Barbosa Nonino
- Division of Nutrition and Metabolism, Department of Health Sciences, Ribeirao Preto Medical School, University of São Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Sao Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Sao Paulo, Brazil
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, New York, USA
| | - Ellen Cristini de Freitas
- School of Physical Education and Sport of Ribeirao Preto (EEFERP/USP), University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, Sao Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Sao Paulo, Brazil
| |
Collapse
|
12
|
Wang D, Zhang M, Zhang Y, Yin Z, Zhang S, Zhao Z, Duan Y. Hepatoprotective effects of polysaccharide from Morchella esculenta are associated with activation of the AMPK/Sirt1 signaling pathway in mice with NAFLD. Int J Biol Macromol 2025; 301:140444. [PMID: 39884630 DOI: 10.1016/j.ijbiomac.2025.140444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The functional food application of edible fungus polysaccharides has been widely studied based on their variety of potential pharmacological activities. However, the hepatoprotective effects and mechanisms of Morchella esculenta polysaccharide against nonalcoholic fatty liver disease (NAFLD) remain unknown. A high-fat diet (HFD) fed C57BL/6 J mice for 8 weeks was employed to establish NAFLD with simple steatosis, methionine choline deficiency (MCD) diet for 4 weeks induced hepatic steatohepatitis and fibrosis. The M. esculenta polysaccharide (MCP) or saline was administered intragastrically. MCP markedly reduced hepatic and serum triglyceride (TG) and cholesterol contents in HFD-fed mice. Moreover, treatment with MCP ameliorated nonalcoholic steatohepatitis (NASH) progression in MCD-fed mice, as evidenced by ameliorated hepatic steatosis, inflammatory response, and fibrosis. Mechanistically, MCP suppressed the expression of lipogenic genes and inflammatory cytokines and upregulated peroxisome proliferator-activated receptor (PPAR)-α expression to induce fatty acid β-oxidation. These beneficial effects were attributed to activating the AMP-activated kinase (AMPK)/Sirtuin 1 (Sirt1) signaling pathway. Therefore, we provided evidence that MCP might be an effective dietary supplement to ameliorate NAFLD.
Collapse
Affiliation(s)
- Dandan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China; College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230601, China
| | - Menglian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China
| | - Yaowen Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China
| | - Zequn Yin
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shuang Zhang
- College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230601, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Yajun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
13
|
Cannella R, Agnello F, Porrello G, Spinello AU, Infantino G, Pennisi G, Cabibi D, Petta S, Bartolotta TV. Performance of ultrasound-guided attenuation parameter and 2D shear wave elastography in patients with metabolic dysfunction-associated steatotic liver disease. Eur Radiol 2025; 35:2339-2350. [PMID: 39373742 PMCID: PMC11914239 DOI: 10.1007/s00330-024-11076-w] [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: 12/29/2023] [Revised: 07/06/2024] [Accepted: 08/23/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE To assess the performance and the reproducibility of ultrasound-guided attenuation parameter (UGAP) and two-dimensional shear wave elastography (2D-SWE) in patients with biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS This study included consecutive adult patients with MASLD who underwent ultrasound with UGAP, 2D-SWE and percutaneous liver biopsy. The median values of 12 consecutive UGAP measurements were acquired by two independent radiologists (R1 and R2). Hepatic steatosis was graded by liver biopsy as: (0) < 5%; (1) 5-33%; (2) > 33-66%; (3) > 66%. Areas under the curve (AUCs) were calculated to determine the diagnostic performance. Inter- and intra-observer reliability was assessed with intraclass correlation coefficient (ICC). RESULTS A hundred patients (median age 55.0 years old) with MASLD were prospectively enrolled. At histopathology, 70 and 42 patients had grade ≥ 2 and 3 steatosis, respectively. Median UGAP was 0.78 dB/cm/MHz (IQR/Med: 5.55%). For the diagnosis of grade ≥ 2 steatosis, the AUCs of UGAP were 0.828 (95% CI: 0.739, 0.896) for R1 and 0.779 (95% CI: 0.685, 0.856) for R2. The inter- and intra-operator reliability of UGAP were excellent, with an ICC of 0.92 (95% CI: 0.87-0.95) and 0.95 (95% CI: 0.92-0.96), respectively. The median liver stiffness was 6.76 kPa (IQR/Med: 16.30%). For the diagnosis of advanced fibrosis, 2D-SWE had an AUC of 0.862 (95% CI: 0.757, 0.934), and the optimal cutoff value was > 6.75 kPa with a sensitivity of 80.6% and a specificity of 75.7%. CONCLUSION UGAP and 2D-SWE provide a good performance for the staging of steatosis and fibrosis in patients with MASLD with an excellent intra-operator reliability of UGAP. KEY POINTS Question How well do ultrasound-guided attenuation parameter (UGAP) and two-dimensional shear wave elastography (2D-SWE) perform for quantifying hepatic steatosis and fibrosis? Findings UGAP had a maximum AUC of 0.828 for the diagnosis of grade ≥ 2 steatosis, and 2D-SWE had an AUC of 0.862 for diagnosing advanced fibrosis. Clinical relevance UGAP and 2D-SWE allow rapid, reproducible, and accurate quantification of hepatic steatosis and fibrosis that can be used for the noninvasive assessment of patients with metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy.
| | - Francesco Agnello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Giorgia Porrello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Alessandro Umberto Spinello
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| | - Giuseppe Infantino
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Grazia Pennisi
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Daniela Cabibi
- Unit of Anatomic Pathology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Tommaso Vincenzo Bartolotta
- Section of Radiology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Via del Vespro 129, Palermo, 90127, Italy
| |
Collapse
|
14
|
Hanamatsu H, Suda G, Ohara M, Ogawa K, Tamaki N, Hikita H, Haga H, Maekawa S, Sugiyama M, Kakisaka T, Nakai M, Sho T, Miura N, Kurosaki M, Asahina Y, Taketomi A, Ueno Y, Takehara T, Nishikaze T, Furukawa JI, Sakamoto N. Elevated A2F bisect N-glycans of serum IgA reflect progression of liver fibrosis in patients with MASLD. J Gastroenterol 2025; 60:456-468. [PMID: 39849179 PMCID: PMC11922979 DOI: 10.1007/s00535-024-02206-8] [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: 07/29/2024] [Accepted: 12/25/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Advanced liver fibrosis in cases of metabolic dysfunction-associated steatotic liver disease (MASLD) leads to cirrhosis and hepatocellular carcinoma. The current gold standard for liver fibrosis is invasive liver biopsy. Therefore, a less invasive biomarker that accurately reflects the stage of liver fibrosis is highly desirable. METHODS This study enrolled 269 patients with liver biopsy-proven MASLD. Patients were divided into three groups (F0/1 (n = 41/85), F2 (n = 47), and F3/4 (n = 72/24)) according to fibrosis stage. We performed serum N-glycomics and identified glycan biomarker for fibrosis stage. Moreover, we explored the carrier proteins and developed a sandwich ELISA to measure N-glycosylation changes of carrier protein. RESULTS Comprehensive N-glycomic analysis revealed significant changes in the expression of A2F bisect and its precursors as fibrosis progressed. The sum of neutral N-glycans carrying bisecting GlcNAc and core Fuc (neutral sum) had a better diagnostic performance to evaluate advanced liver fibrosis (AUC = 0.804) than conventional parameters (FIB4 index, aspartate aminotransferase-to-alanine aminotransferase ratio (AAR), and serum level of Mac-2-binding protein glycol isomer (M2BPGi). The combination of the neutral sum and FIB4 index enhanced diagnostic performance (AUC = 0.840). IgM, IgA, and complement C3 were identified as carrier proteins with A2F bisect N-glycan. A sandwich ELISA based on N-glycans carrying bisecting GlcNAc and IgA showed similar diagnostic performance than the neutral sum. CONCLUSIONS A2F bisect N-glycan and its precursors are promising candidate biomarkers for advanced fibrosis in MASLD patients. Analysis of these glycan alterations on IgA may have the potential to serve as a novel ELISA diagnostic tool for MASLD in routine clinical practice. CLINICAL TRIAL NUMBER UMIN000030720.
Collapse
Affiliation(s)
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Masatsugu Ohara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Koji Ogawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Nobuharu Tamaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Haga
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Shinya Maekawa
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Masaya Sugiyama
- Department of Viral Pathogenesis and Controls, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tatsuhiko Kakisaka
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Masato Nakai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Takuya Sho
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Nobuaki Miura
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Aichi, Japan
| | - Masayuki Kurosaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Yoshiyuki Ueno
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takashi Nishikaze
- Solutions COE, Analytical and Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Jun-Ichi Furukawa
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Aichi, Japan.
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan.
| |
Collapse
|
15
|
Koch V, Gotta J, Chernyak V, Cengiz D, Torgashov K, Eichler K, Vilgrain V, Martin SS, Ziegengeist NS, Konrad P, Booz C, Yel I, D'Angelo T, Mahmoudi S, Scholtz J, Bernatz S, Alizadeh LS, Cimprich M, Solim LA, Thalhammer A, Gruber‐Rouh T, Hammerstingl RM, Zeuzem S, Finkelmeier F, Pathil‐Warth A, Onay M, Kinzler MN, Darwish O, Annio G, Taylor SA, Wild P, Dahmer I, Herrmann E, Almansour H, Vogl TJ, Gruenewald LD, Sinkus R. Biomechanical Assessment of Liver Integrity: Prospective Evaluation of Mechanical Versus Acoustic MR Elastography. J Magn Reson Imaging 2025; 61:1890-1904. [PMID: 39165139 PMCID: PMC11896941 DOI: 10.1002/jmri.29560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Magnetic resonance elastography (MRE) can quantify tissue biomechanics noninvasively, including pathological hepatic states like metabolic dysfunction-associated steatohepatitis. PURPOSE To compare the performance of 2D/3D-MRE using the gravitational (GT) transducer concept with the current commercial acoustic (AC) solution utilizing a 2D-MRE approach. Additionally, quality index markers (QIs) were proposed to identify image pixels with sufficient quality for reliably estimating tissue biomechanics. STUDY TYPE Prospective. POPULATION One hundred seventy participants with suspected or confirmed liver disease (median age, 57 years [interquartile range (IQR), 46-65]; 66 females), and 11 healthy volunteers (median age, 31 years [IQR, 27-34]; 5 females). FIELD STRENGTH/SEQUENCE Participants were scanned twice at 1.5 T and 60 Hz vibration frequency: first, using AC-MRE (2D-MRE, spin-echo EPI sequence, 11 seconds breath-hold), and second, using GT-MRE (2D- and 3D-MRE, gradient-echo sequence, 14 seconds breath-hold). ASSESSMENT Image analysis was performed by four independent radiologists and one biomedical engineer. Additionally, superimposed analytic plane shear waves of known wavelength and attenuation at fixed shear modulus were used to propose pertinent QIs. STATISTICAL TESTS Spearman's correlation coefficient (r) was applied to assess the correlation between modalities. Interreader reproducibility was evaluated using Bland-Altman bias and reproducibility coefficients. P-values <0.05 were considered statistically significant. RESULTS Liver stiffness quantified via GT-2D/3D correlated well with AC-2D (r ≥ 0.89 [95% CI: 0.85-0.92]) and histopathological grading (r ≥ 0.84 [95% CI: 0.72-0.91]), demonstrating excellent agreement in Bland-Altman plots and between readers (κ ≥ 0.86 [95% CI: 0.81-0.91]). However, GT-2D showed a bias in overestimating stiffness compared to GT-3D. Proposed QIs enabled the identification of pixels deviating beyond 10% from true stiffness based on a combination of total wave amplitude, temporal sinusoidal nonlinearity, and wave signal-to-noise ratio for GT-3D. CONCLUSION GT-MRE represents an alternative to AC-MRE for noninvasive liver tissue characterization. Both GT-2D and 3D approaches correlated strongly with the established commercial approach, offering advanced capabilities in abdominal imaging compared to AC-MRE. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Vitali Koch
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Jennifer Gotta
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Victoria Chernyak
- Department of RadiologyMemorial Sloan Kettering Cancer CenterNew York CityNew YorkUSA
| | - Duygu Cengiz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Katerina Torgashov
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Katrin Eichler
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Valérie Vilgrain
- Laboratory of Imaging Biomarkers, Center for Research on Inflammation, UMR 1149 INSERM, Université de ParisParisFrance
- Department of RadiologyBeaujon University Hospital Paris Nord, AP‐HPClichyFrance
| | - Simon S. Martin
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Nicole S. Ziegengeist
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Paul Konrad
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Christian Booz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Ibrahim Yel
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Tommaso D'Angelo
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Scherwin Mahmoudi
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Jan‐Erik Scholtz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Simon Bernatz
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Leona S. Alizadeh
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Marina Cimprich
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Levent A. Solim
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Axel Thalhammer
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Tatjana Gruber‐Rouh
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Renate M. Hammerstingl
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Stefan Zeuzem
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Fabian Finkelmeier
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Anita Pathil‐Warth
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Melis Onay
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Maximilian N. Kinzler
- Department of Internal Medicine IGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Omar Darwish
- School of Biomedical Engineering and Imaging Sciences, King's College LondonLondonUK
| | - Giacomo Annio
- Laboratory of Translational Vascular Sciences, U1148, INSERM, Université de ParisParisFrance
| | - Stuart A. Taylor
- Radiology DepartmentUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Peter Wild
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Iulia Dahmer
- Institute of Biostatistics and Mathematical Modeling, Faculty of Medicine, Goethe University FrankfurtFrankfurt am MainGermany
| | - Eva Herrmann
- Institute of Biostatistics and Mathematical Modeling, Faculty of Medicine, Goethe University FrankfurtFrankfurt am MainGermany
| | - Haidara Almansour
- Department of Diagnostic and Interventional RadiologyEberhard‐Karls University TuebingenTuebingenGermany
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Leon D. Gruenewald
- Department of Diagnostic and Interventional RadiologyGoethe University Frankfurt, University HospitalFrankfurt am MainGermany
| | - Ralph Sinkus
- School of Biomedical Engineering and Imaging Sciences, King's College LondonLondonUK
- Laboratory of Translational Vascular Sciences, U1148, INSERM, Université de ParisParisFrance
| |
Collapse
|
16
|
Lefere S, Mosca A, Hudert C, Dupont E, Fitzpatrick E, Kyrana E, Dhawan A, Kalveram L, Pietrobattista A, Geerts A, De Bruyne R. Development and validation of pFIB scores for exclusion of significant liver fibrosis in pediatric MASLD. Hepatology 2025; 81:1276-1287. [PMID: 39028885 DOI: 10.1097/hep.0000000000001016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/01/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent pediatric liver disease, yet accurate risk scores for referral of children/adolescents with suspected clinically significant liver fibrosis are currently lacking. APPROACH AND RESULTS Clinical and biochemical variables were collected in a prospective cohort of 327 children and adolescents with severe obesity, in whom liver fibrosis was evaluated by transient elastography. Logistic regression was performed to establish continuous (pFIB-c) and simplified (pFIB-6) diagnostic scores that accurately exclude significant (≥F2) fibrosis. Performance for each was compared to established noninvve fibrosis scores. These scores were validated in elastography (n=504) and multiple biopsy-proven MASLD (n=261) cohorts. Patient sex, ethnicity, weight z-score, homeostatic model assessment of insulin resistance index, ALT, and presence of hypertension were included in the scores. The pFIB-c and pFIB-6 exhibited good discriminatory capacity (c-statistic of 0.839 and 0.826), outperforming existing indices. Negative predictive values were >90% for both scores in the derivation and elastography validation cohorts. Performance in the histological cohorts varied (AUROCs for the pFIB-c between 0.710 and 0.770), as the scores were less accurate when applied to populations in tertiary referral centers characterized by a high prevalence of significant fibrosis and high ALT levels. CONCLUSIONS Analyzing several cohorts totaling approximately 1100 children and adolescents, we developed novel risk scores incorporating readily available clinical variables. In accordance with the aim of excluding pediatric MASLD-associated fibrosis, the scores performed better in nonselected cohorts of children and adolescents living with obesity than in patients referred to tertiary liver units.
Collapse
Affiliation(s)
- Sander Lefere
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University Hospital, Ghent, Belgium
| | - Antonella Mosca
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Christian Hudert
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Emer Fitzpatrick
- Paediatric Liver, GI and Nutrition Centre, King's College Hospital, London, United Kingdom
- Department of Gastroenterology, Hepatology and Nutrition, Children's Health Ireland and University College Dublin, Ireland
| | - Eirini Kyrana
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Anil Dhawan
- Paediatric Liver, GI and Nutrition Centre, King's College Hospital, London, United Kingdom
| | - Laura Kalveram
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Pietrobattista
- Hepatogastroenterology, Nutrition, Digestive Endoscopy and Liver Transplant Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University Hospital, Ghent, Belgium
| | - Ruth De Bruyne
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Ghent University, Ghent, Belgium
| |
Collapse
|
17
|
Elkattawy HA, Alsemeh AE, Ali LS, Ahmed MM, Eltaweel AM, Shaikh FM, Behiry A, Hassan AES, Sabir DK, Elsherbini DMA, Ali SK, Zakari MO, Mojaddidi MA, Ali EK, Elbastawisy YM, Hadhoud S. Decorin alleviates non-alcoholic fatty liver disease in rats with polycystic ovary syndrome. Tissue Cell 2025; 93:102689. [PMID: 39705869 DOI: 10.1016/j.tice.2024.102689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/09/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Endocrine multisystem defect polycystic ovary syndrome (PCOS) causes hyperandrogenism and infertility. Half of PCOS women have (non-alcoholic fatty liver disease) NAFLD, which increases metabolic disease risk. We tested decorin's effect on NAFLD and related processes in PCOS. NAFLD+PCOS, PCOS+decorin, and control rats were studied. Decorin was evaluated on NAFLD/PCOS rats. Test group rats received HF for eight weeks to generate NAFLD. The rats got 1 mg/kg letrozole orally daily for 21 days to diagnosis PCOS. Afterward, rats got injectable decorin for 14 days. Body weight, liver weight, liver coefficient Abdominal Circumference (AC) and body mass index (BMI) were determined. Blood triglycerides (TG), total cholesterol, LDL-c, AST, and glucose were measured. The insulin, testosterone, estrogen, LH, and FSH were measured by ELISA. GPx, SOD, MDA, TNF-, and Caspase-3 liver immunohistochemistry were evaluated. NAFLD liver tissues in PCOS models showed biochemical and histological alterations. NAFLD+PCOS raised BMI, AC, liver weight, and coefficient. Blood glucose, insulin resistance, TG, ALT, and AST increased. Lipid abnormalities (TG, cholesterol, LDL-c, and HDL-c), oxidative stress markers (MDA, SOD, and GPx), and liver dysfunction were found. Low serum E2 and high T supported PCO. Decorin reduced model rat BMI, liver weight, coefficient, insulin resistance, TG, ALT, and AST. It reduced liver inflammation, improved liver extract lipids, and normalized MDA, SOD, and GPx. In the model group, decorin lowered serum T, E2, LH, caspase 3, and TNF-alpha. Decorating improved NAFLD/PCOS group liver histology and function. Decorin reduces hepatosteatosis by reducing liver inflammation, oxidative stress, and dyslipidemia.
Collapse
Affiliation(s)
- Hany A Elkattawy
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah 13713, Saudi Arabia.
| | - Amira Ebrahim Alsemeh
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Lashin Saad Ali
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, P.O. Box 19328, Amman, Jordan; Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Mona Mostafa Ahmed
- Pathology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Asmaa Monir Eltaweel
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt; College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh 11481, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia.
| | - Farha M Shaikh
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah 13713, Saudi Arabia.
| | - Ahmed Behiry
- Department of Tropical Medicine and Endemic Diseases, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Ahmed El-Sayed Hassan
- Medical Physiology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt; Department of Basic Medical Sciences, College of Medicine, Sulaiman Al-Rajhi University, Bukayriah 51941, Saudi Arabia.
| | - Deema Kamal Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Dalia Mahmoud Abdelmonem Elsherbini
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia; Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Sahar K Ali
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah 13713, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Madaniah Omar Zakari
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Medina, Saudi Arabia.
| | - Moaz Abdullah Mojaddidi
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Medina, Saudi Arabia.
| | - Ehab Kamal Ali
- Department of Anatomy and Embryology, Faculty of Medicine, AL-Azhar University, New Damietta, Egypt.
| | - Yasser M Elbastawisy
- Department of Basic Medical Sciences, College of Medicine, Al-Rayan Colleges, Al-Madinah, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Shimaa Hadhoud
- Medical Physiology Department, Faculty of Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| |
Collapse
|
18
|
Azizi N, Naghibi H, Shakiba M, Morsali M, Zarei D, Abbastabar H, Ghanaati H. Evaluation of MRI proton density fat fraction in hepatic steatosis: a systematic review and meta-analysis. Eur Radiol 2025; 35:1794-1807. [PMID: 39254718 DOI: 10.1007/s00330-024-11001-1] [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: 02/10/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Amidst the global rise of metabolic dysfunction-associated steatotic liver disease (MASLD), driven by increasing obesity rates, there is a pressing need for precise, non-invasive diagnostic tools. Our research aims to validate MRI Proton Density Fat Fraction (MRI-PDFF) utility, compared to liver biopsy, in grading hepatic steatosis in MASLD. METHODS A systematic search was conducted across Embase, PubMed/Medline, Scopus, and Web of Science until January 13, 2024, selecting studies that compare MRI-PDFF with liver biopsy for hepatic steatosis grading, defined as grades 0 (< 5% steatosis), 1 (5-33% steatosis), 2 (34-66% steatosis), and 3 (> 66% steatosis). RESULTS Twenty-two studies with 2844 patients were included. The analysis showed high accuracy of MRI-PDFF with AUCs of 0.97 (95% CI = 0.96-0.98) for grade 0 vs ≥ 1, 0.91 (95% CI = 0.88-0.93) for ≤ 1 vs ≥ 2, and 0.91 (95% CI = 0.88-0.93) for ≤ 2 vs 3, diagnostic odds ratio (DOR) from 98.74 (95% CI = 58.61-166.33) to 23.36 (95% CI = 13.76-39.68), sensitivity and specificity from 0.93 (95% CI = 0.88-0.96) to 0.76 (95% CI = 0.63-0.85) and 0.93 (95% CI = 0.88-0.96) to 0.89 (95% CI = 0.84-0.93), respectively. Likelihood ratio (LR) + ranged from 13.3 (95% CI = 7.4-24.0) to 7.2 (95% CI = 4.9-10.5), and LR - from 0.08 (95% CI = 0.05-0.13) to 0.27 (95% CI = 0.17-0.42). The proposed MRI-PDFF threshold of 5.7% for liver fat content emerges as a potential cut-off for the discrimination between grade 0 vs ≥ 1 (p = 0.075). CONCLUSION MRI-PDFF is a precise non-invasive technique for diagnosing and grading hepatic steatosis, warranting further studies to establish its diagnostic thresholds. CLINICAL RELEVANCE STATEMENT This study underscores the high diagnostic accuracy of MRI-PDFF for distinguishing between various grades of hepatic steatosis for early detection and management of MASLD, though further research is necessary for broader application. KEY POINTS MRI-PDFF offers precision in diagnosing and monitoring hepatic steatosis. The diagnostic accuracy of MRI-PDFF decreases as the grade of hepatic steatosis advances. A 5.7% MRI-PDFF threshold differentiates steatotic from non-steatotic livers.
Collapse
Affiliation(s)
- Narges Azizi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Hamed Naghibi
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Madjid Shakiba
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Mina Morsali
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Diana Zarei
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Hedayat Abbastabar
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran
| | - Hossein Ghanaati
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
19
|
Deng J, Ding K, Liu S, Chen F, Huang R, Xu B, Zhang X, Xie W. SOX9 Overexpression Ameliorates Metabolic Dysfunction-associated Steatohepatitis Through Activation of the AMPK Pathway. J Clin Transl Hepatol 2025; 13:189-199. [PMID: 40078197 PMCID: PMC11894392 DOI: 10.14218/jcth.2024.00197] [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: 06/14/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025] Open
Abstract
Background and Aims The transcription factor sex-determining region Y-related high-mobility group-box gene 9 (SOX9) plays a critical role in organ development. Although SOX9 has been implicated in regulating lipid metabolism in vitro, its specific role in metabolic dysfunction-associated steatohepatitis (MASH) remains poorly understood. This study aimed to investigate the role of SOX9 in MASH pathogenesis and explored the underlying mechanisms. Methods MASH models were established using mice fed either a methionine- and choline-deficient (MCD) diet or a high-fat, high-fructose diet. To evaluate the effects of SOX9, hepatocyte-specific SOX9 deletion or overexpression was performed. Lipidomic analyses were conducted to assess how SOX9 influences hepatic lipid metabolism. RNA sequencing was employed to identify pathways modulated by SOX9 during MASH progression. To elucidate the mechanism further, HepG2 cells were treated with an adenosine monophosphate-activated protein kinase (AMPK) inhibitor to test whether SOX9 acts via AMPK activation. Results SOX9 expression was significantly elevated in hepatocytes of MASH mice. Hepatocyte-specific SOX9 deletion exacerbated MCD-induced MASH, whereas overexpression of SOX9 mitigated high-fat, high-fructose-induced MASH. Lipidomic and RNA sequencing analyses revealed that SOX9 suppresses the expression of genes associated with lipid metabolism, inflammation, and fibrosis in MCD-fed mice. Furthermore, SOX9 deletion inhibited AMPK pathway activation, while SOX9 overexpression enhanced it. Notably, administration of an AMPK inhibitor negated the protective effects of SOX9 overexpression, leading to increased lipid accumulation in HepG2 cells. Conclusions Our findings demonstrate that SOX9 overexpression alleviates hepatic lipid accumulation in MASH by activating the AMPK pathway. These results highlight SOX9 as a promising therapeutic target for treating MASH.
Collapse
Affiliation(s)
- Juan Deng
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shuqing Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fei Chen
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ru Huang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bonan Xu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weifen Xie
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| |
Collapse
|
20
|
Yoon J, Choi WI, Lee WH, Lee GB, Choi BW, Kim P, Heo Y, Kim DG, Kim HA, Bae MA, Kim SS, Lee EY, Oh CM, Lee HJ, Kim HW, Namkung W, Kim H, Ahn JH. Synthesis and Biological Evaluation of Peripheral 5HT 2B Antagonists for Liver Fibrosis. J Med Chem 2025; 68:6493-6506. [PMID: 40048549 DOI: 10.1021/acs.jmedchem.4c03003] [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: 03/28/2025]
Abstract
Liver fibrosis is characterized by an excessive accumulation of extracellular matrix components, leading to the distortion of liver architecture and function. Recent studies have shown that antagonizing 5-hydroxytryptamine receptor 2B (5HT2B) stimulates the apoptosis of activated hepatic stellate cells and inhibits their proliferation while concurrently regressing hepatocyte proliferation. In this study, we present compound 19c, which demonstrates promising efficacy both in vitro and in vivo. 19c showed robust in vitro activity with an IC50 value of 1.09 nM and limited blood-brain barrier penetration. Furthermore, 19c did not significantly inhibit hERG and cytochrome P450 enzymes. 19c markedly reduced fibrotic deposition, with a decrease in fibrosis stage and area in the CCl4-induced liver fibrosis mouse model. Additionally, treatment with 19c led to downregulation of key fibrosis-related genes, including α-SMA, Timp1, Col1a1, and Col3a1. Taken together, these results suggest that 19c has the potential to be a novel antifibrotic agent.
Collapse
Affiliation(s)
- Jihyeon Yoon
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Won-Il Choi
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Won Hee Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Gwi Bin Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Byeong Wook Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Pyeongkeun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yerim Heo
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Dong Gun Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyeon Ah Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Seong Soon Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Eun Young Lee
- JD Bioscience Inc., TJS Knowledge Industrial Center, Suite 801, 208 Beon-gil Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyeok Jae Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyun Woo Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- Center for Quantum Information, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Wan Namkung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Biomedical Research Center, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- JD Bioscience Inc., TJS Knowledge Industrial Center, Suite 801, 208 Beon-gil Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| |
Collapse
|
21
|
Truong E, Alnimer L, Gornbein JA, Yang JD, Alkhouri N, Harrison SA, Noureddin M. Agile 3+ and 4 Scores Accurately Predict Major Adverse Liver Outcomes, Liver Transplant, Progression of MELD Score, the Development of Hepatocellular Carcinoma, and Death in NAFLD. Dig Dis Sci 2025:10.1007/s10620-025-08850-1. [PMID: 40126753 DOI: 10.1007/s10620-025-08850-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 01/04/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND AND AIMS Based on liver stiffness measurement by vibration controlled transient elastography (LSM by VCTE), the Agile 3+ and 4 are novel noninvasive scores that accurately identify advanced fibrosis (≥ F3) and cirrhosis (F4), respectively. We investigated and compared the Agile 3+ and 4 scores' performances in predicting adverse events to LSM alone, FIB-4 and Fibroscan-AST (FAST) score. METHOD This retrospective analysis included NAFLD patients with LSM by VCTE and laboratory testing from a tertiary care center from 2013 to 2022. Adverse events were defined as major adverse liver outcomes (MALO), hepatocellular carcinoma, liver transplant, and death. MALO was defined as ascites, hepatic encephalopathy, or esophageal variceal bleeding. We used the Cox proportional hazard rate model and the Harrell's concordance (C) statistic to compare predictive performances. RESULTS 733 total subjects with median follow-up of 27.0 months were included. Average age was 58.1 years and 32.8% had type 2 diabetes. Average alanine aminotransferase was 46.6 IU/L, aspartate aminotransferase: 34.5 IU/L, albumin: 4.4 g/dL, and platelets: 241.1 × 109/L. Fourteen subjects had 21 adverse outcomes, including 10 MALO, 5 HCC, 4 liver transplants, 3 progression of MELD score, and 6 deaths. Agile 3+ and 4 respectively had the highest C stats of 0.911 (C stat SE 0.028) and 0.909 (C stat SE 0.029) compared to LSM (C stat 0.857, C stat SE 0.045), FIB-4 (C stat 0.843, C stat SE 0.037) or FAST (C stat 0.703, C stat SE 0.085). CONCLUSION The Agile 3+ and 4 scores had the highest likelihood of accurately predicting adverse outcomes including MALO and death compared to LSM alone, FIB-4 or FAST score.
Collapse
Affiliation(s)
- Emily Truong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lynna Alnimer
- Division of Gastroenterology, Henry Ford Providence Hospital, Michigan State University/College of Human Medicine, Southfield, MI, USA
| | - Jeffrey A Gornbein
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Comprehensive Transplant Center, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | | | | | - Mazen Noureddin
- Houston Methodist Hospital, Houston Research Institute, 1155 Dairy Ashford Suite 200, Houston, TX, 77079, USA.
- Lynda K. and David M. Underwood Center for Digestive Disorders, Department of Medicine, J.C. Walter Jr. Transplant Center, Sherrie & Alan Conover Center for Liver Disease & Transplantation, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, USA.
| |
Collapse
|
22
|
Wu Y, Han Y, Zheng L, Liu L, Li W, Zhang F. Validation of the diagnostic accuracy of the acFibroMASH index for at-risk MASH in patients with metabolic dysfunction-associated steatotic liver disease. BMC Gastroenterol 2025; 25:196. [PMID: 40128689 PMCID: PMC11931867 DOI: 10.1186/s12876-025-03781-7] [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: 11/05/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
OBJECTIVE The objective of this study was to validate the diagnostic accuracy of the acFibroMASH index in a population of metabolic dysfunction-associated steatotic liver disease (MASLD) patients with at-risk metabolic dysfunction-associated steatohepatitis (MASH) and to compare it with other scoring systems. METHODS 394 patients with biopsy-proven MASLD were retrospectively enrolled. The patients were divided into the at-risk MASH (NAFLD activity score ≥ 4 and significant fibrosis) group (n = 103) and the non-at-risk MASH group (n = 291). The diagnostic performance of the acFibroMASH index was compared to that of fibroScan-aspartate aminotransferase (FAST) and other noninvasive fibrosis scores by plotting the receiver operating characteristic curve (ROC), including the area under the curve (AUC), sensitivity, and specificity. Cut-offs of the acFibroMASH index for sensitivity (≥ 0.90) and specificity (≥ 0.90) were obtained in our cohort. RESULTS The AUC of the acFibroMASH index in assessing at-risk MASH was 0.780, while the AUC of FAST was 0.770. The comparison of acFibroMASH with FAST showed no significant difference (P = 0.542). When the cut-off value for acFibroMASH was < 0.15, 95.5% of at-risk MASH patients could be excluded in 89 patients correctly. Conversely, when the cut-off value was set at > 0.39, 49.3% of at-risk MASH patients could be diagnosed in 140 patients correctly. When the NPV was set at 0.900, the critical value for exclusion was determined to be 0.23, with a sensitivity of 0.835 and a specificity of 0.526. CONCLUSION This study validated the efficacy of the acFibroMASH index in predicting at-risk MASH in a population of MASLD patients, demonstrating comparable performance to that of the FAST. The acFibroMASH index may provide a valuable clinical basis for screening and identifying at-risk MASH in primary care settings.
Collapse
Affiliation(s)
- Yunfei Wu
- Department of Pathology, Changzhou Third People's Hospital, Changzhou, 213001, China
| | - Yan Han
- Department of Endocrinology, Changzhou Third People's Hospital, Changzhou, 213001, China
- Department of Clinical Nutrition, Changzhou Third People's Hospital, Changzhou, 213001, China
| | - Liming Zheng
- Clinical Laboratory, Changzhou Third People's Hospital, Changzhou, 213001, China
| | - Longgen Liu
- Department of Liver Diseases, Changzhou Third People's Hospital, Changzhou, 213001, China
| | - Wenjian Li
- Department of Urology, Changzhou Third People's Hospital, Changzhou, 213001, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213001, China.
| | - Fan Zhang
- Department of Endocrinology, Changzhou Third People's Hospital, Changzhou, 213001, China.
- Department of Clinical Nutrition, Changzhou Third People's Hospital, Changzhou, 213001, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213001, China.
| |
Collapse
|
23
|
Sun X, Yang Z, Min L, Gong S, Miao X, Wang B, Kong X, Zhu Q. Interferon regulatory factor 1 contributes to metabolic dysfunction associated steatotic liver disease. Life Sci 2025:123575. [PMID: 40132726 DOI: 10.1016/j.lfs.2025.123575] [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: 02/03/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
AIMS Non-alcoholic fatty liver disease (NAFLD) or metabolic dysfunction associated steatotic liver disease (MASLD), has reached epidemic levels in multiple regions worldwide and contributes to cirrhosis and hepatocellular carcinoma. We have previously reported that the CC motif chemokine ligand 11 (CCL11) is a key regulator of NAFLD and expression of interferon regulatory factor 1 (IRF1) can be up-regulated by CCL11 in hepatocytes, the relevance of which is not clear. In the present study we investigated the role of IRF1 in NAFLD pathogenesis. METHODS AND MATERIALS NAFLD was investigated in mice fed a high-fat high carbohydrate (HFHC) diet or in the genetically predisposed obese mice (db/db). KEY FINDINGS Hepatocytes from CCL11 knockout mice displayed a less severe NAFLD phenotype, when treated with palmitic acid (PA), compared to wild type hepatocytes, which could be normalized by IRF1 over-expression. On the contrary, IRF1 knockdown in hepatocytes significantly down-regulated expression of pro-inflammatory mediators and dampened lipid accumulation induced by PA treatment. More importantly, IRF1 knockdown in mice led to amelioration of NAFLD in mice. RNA-seq and CUT&Tag-seq identified pro-NAFLD genes, including Osbpl3, Ddit4, and Ccl2, as potential targets for IRF1 in hepatocytes. SIGNIFICANCE Our data reveal a novel regulatory role of IRF1 in NAFLD pathogenesis. Targeting IRF1 can be considered as a reasonable approach for NAFLD intervention.
Collapse
Affiliation(s)
- Xinyue Sun
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Zhen Yang
- Graduate School of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Li Min
- Center for Experimental Medicine, Department of Pathophysiology, Jiangsu Health Vocational College, China
| | - Shanwen Gong
- Institute of Biomedical Research, College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Xiulian Miao
- Institute of Biomedical Research, College of Agriculture and Biology, Liaocheng University, Liaocheng, China
| | - Bo Wang
- Department of General Surgery, Wujin Affiliated Hospital of Jiangsu University, the Wujin Clinical College of Xuzhou Medical University, Changzhou, China.
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Qiang Zhu
- Department of Liver Transplantation, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
24
|
Jönsson C, Ma'ayeh S, Zhang B, Kechagias S, Liljeblad M, Nasr P, Hansson SF, Ekstedt M. Vascular endothelial growth factor A, a potential non-invasive biomarker for metabolic dysfunction-associated steatotic liver disease progression. Clin Biochem 2025; 137:110920. [PMID: 40127834 DOI: 10.1016/j.clinbiochem.2025.110920] [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: 09/09/2024] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
INTRODUCTION AND OBJECTIVES Liver fibrosis is the primary predictor of complications in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). However, there are currently no non-invasive prognostic tests to stratify patients at risk for hepatic fibrosis progression. This study aimed to explore whether plasma proteins could serve as non-invasive biomarkers for monitoring MASLD disease progression. MATERIALS AND METHODS Blood plasma protein analysis was performed on samples from a long-term follow-up study of patients with MASLD with repeated liver biopsies. Over 1100 proteins covering a broad range of biological processes were analyzed using 13 Olink® Target 96 panels. Protein level changes were compared between the different time points and between patients with or without an increase in the liver fibrosis stage between the two biopsies. RESULTS Increased vascular endothelial growth factor A (VEGFA) plasma levels were significantly associated with liver fibrosis progression in patients with a histologically assessed increase in the fibrosis stage. CONCLUSIONS These findings suggest that the plasma protein VEGFA may be an effective biomarker for monitoring fibrosis progression in patients with MASLD.
Collapse
Affiliation(s)
- Cecilia Jönsson
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden.
| | | | | | - Stergios Kechagias
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden.
| | - Mathias Liljeblad
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden.
| | - Patrik Nasr
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden; Wallenberg Centre for Molecular Medicine, Linköping University, Sweden.
| | - Sara F Hansson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden.
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, SE-581 83 Linköping, Sweden.
| |
Collapse
|
25
|
Farzi M, McGenity C, Cratchley A, Leplat L, Bankhead P, Wright A, Treanor D. Liver-Quant: Feature-based image analysis toolkit for automatic quantification of metabolic dysfunction-associated steatotic liver disease. Comput Biol Med 2025; 190:110049. [PMID: 40121800 DOI: 10.1016/j.compbiomed.2025.110049] [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: 10/18/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Liver biopsy assessment by pathologists remains the gold standard for diagnosing metabolic dysfunction-associated steatotic liver disease (MASLD). Current automated image analysis tools for patient risk stratification are often proprietary or not applicable to whole slide images (WSIs). Here, we introduce "Liver-Quant," an open-source Python package for quantifying steatosis and fibrosis in liver WSIs. METHOD Liver-Quant leverages colour and morphological features to measure Steatosis Proportionate Area (SPA) and Collagen Proportionate Area (CPA). We evaluated the method using an internal dataset of 414 WSIs from adult patients (Leeds Teaching Hospitals NHS Trust, 2016-2022) and an external public dataset (109 WSIs). Semi-quantitative scores were extracted from pathological reports. The Spearman rank coefficient (ρ) assessed correlations between computed SPA/CPA and pathologist scores. RESULTS Steatosis quantification showed a substantial correlation (ρ = 0.92), while fibrosis quantification yielded a moderate correlation (ρ = 0.51). We further investigated the impact of three staining dyes (Van Gieson (VG), Picro Sirius Red (PSR), and Masson's Trichrome (MTC)) on fibrosis quantification (n = 18). Stain normalisation yielded excellent agreement in CPA measurements across all three stains. Without normalisation, PSR achieved the strongest correlation with human scores (ρ = 0.9) followed by VG (ρ = 0.8) and MTC (ρ = 0.59). Finally, we explored the impact of apparent magnification on SPA and CPA. High-resolution images (0.25 or 0.50 μm per pixel (MPP)) were necessary for accurate SPA measurement, while lower resolution (10 MPP) sufficed for CPA measurements. CONCLUSIONS Liver-Quant offers an open-source solution for rapid and precise MASLD quantification in WSIs applicable to multiple histological stains.
Collapse
Affiliation(s)
- Mohsen Farzi
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK; University of Leeds, Leeds, UK.
| | - Clare McGenity
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK; University of Leeds, Leeds, UK
| | - Alyn Cratchley
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Leo Leplat
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Peter Bankhead
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK; Edinburgh Pathology and CRUK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Alexander Wright
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK; University of Leeds, Leeds, UK
| | - Darren Treanor
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK; University of Leeds, Leeds, UK; Department of Clinical Pathology & Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Gu J, Shen Y, Guo L, Chen Z, Zhou D, Ji G, Gu A. Investigation of the mechanisms of liver injury induced by emamectin benzoate exposure at environmental concentrations in zebrafish: A multi-omics approach to explore the role of the gut-liver axis. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:138008. [PMID: 40132265 DOI: 10.1016/j.jhazmat.2025.138008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/27/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Emamectin benzoate (EMB) is a lipophilic pesticide that enters aquatic systems and adversely affects non-target organisms. This study investigated the long-term effects of EMB on zebrafish, exposing them to concentrations of 0, 0.1, 1, and 10 μg/L from the 4-hour post-fertilization (hpf) embryo stage to the 120-day post-fertilisation (dpf) adult stage. We found that exposure to 1 μg/L EMB induced liver damage, manifested as impaired liver function (elevated aspartate aminotransferase (AST) and alanine aminotransferase (ALT)), histopathological damage (lipid accumulation), as well as inflammatory and oxidative damage, with a dose - dependent effect. Non-targeted metabolomic analysis revealed an increase in lipid molecules in the liver, affecting the pathways related to glycerophospholipid metabolism. In addition, EMB exposure resulted in damage to the intestinal barrier and inflammatory responses in zebrafish. 16S rRNA sequencing demonstrated that EMB exposure resulted in notable alterations in the gut microbiota composition. Notably, the abundance of Plesiomonas and Cetobacterium increased in the EMB exposure group and exhibited a positive correlation with the majority of liver lipid metabolites. In contrast, reductions in Muribaculaceae and Alloprevotella were negatively correlated. The results of this study indicate that long-term exposure to EMB disrupts the gut microbiota, leading to the dysregulation of hepatic phospholipid metabolism. These findings provide new insights into the health risks associated with EMB and highlight its potential threats to higher organisms, including mammals.
Collapse
Affiliation(s)
- Jie Gu
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Yuehong Shen
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Liguo Guo
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China
| | - Zhicheng Chen
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology,Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Dingyu Zhou
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Guixiang Ji
- Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment of the People's Republic of China, Nanjing Institute of Environmental science, Ministry of Ecology and Environment, Nanjing 210042, China.
| | - Aihua Gu
- Jiangsu Environmental Health Risk Assessment Engineering Research Center, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
27
|
Wood GC, Hoovler A, Luthra R, Still CD, Shariff H, Still M, Hayes J, Benotti P, Uzoigwe C. Noninvasive identification of metabolic dysfunction-associated steatohepatitis (INFORM MASH): a retrospective cohort and disease modeling study. Expert Rev Gastroenterol Hepatol 2025:1-9. [PMID: 40067340 DOI: 10.1080/17474124.2025.2477249] [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: 12/09/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Using common clinical parameters, we aimed to noninvasively identify and predict metabolic dysfunction-associated steatohepatitis (MASH)/MASH with clinically significant fibrosis. RESEARCH DESIGN AND METHODS Patients aged ≥18 with electronic health record (EHR) documented liver function tests and liver biopsies between 2016 and 2021 were retrospectively identified from the Geisinger Health System Research Liver Registry. MASH cases were confirmed using the nonalcoholic fatty liver disease (NAFLD) activity score. Training and validation datasets were used to create an algorithm/predictive model assessing EHR-derived predictors of MASH/MASH with clinically significant fibrosis (fibrosis stage F2-F4). Predictive accuracy was evaluated using the area under the curve. RESULTS The analysis included 2698 patients. We created a composite likelihood score using variables significant for MASH and/or MASH with clinically significant fibrosis: liver enzymes (alanine aminotransferase [ALT], aspartate aminotransferase [AST]), prior year AST, metabolic disease, pulse (heart rate), and body mass index. The score had higher sensitivity and specificity for predicting MASH than Fibrosis-4 (FIB-4) Index, AST to platelet ratio index (APRI), and NAFLD fibrosis score (NFS); sensitivity and specificity were comparable to FIB-4 and APRI for predicting MASH with clinically significant fibrosis but superior to NFS. CONCLUSION The composite likelihood score could potentially be a tool for early MASH screening.
Collapse
Affiliation(s)
- G Craig Wood
- Geisinger Health System Center for Obesity and Metabolic Research, Danville, PA, USA
| | | | | | - Christopher D Still
- Geisinger Health System Center for Obesity and Metabolic Research, Danville, PA, USA
| | - Hamzah Shariff
- Geisinger Health System Center for Obesity and Metabolic Research, Danville, PA, USA
| | - Matthew Still
- Geisinger Health System Center for Obesity and Metabolic Research, Danville, PA, USA
| | - Jonathan Hayes
- Geisinger Health System Center for Obesity and Metabolic Research, Danville, PA, USA
| | - Peter Benotti
- Geisinger Health System Center for Obesity and Metabolic Research, Danville, PA, USA
| | | |
Collapse
|
28
|
Long L, Wu Y, Tang H, Xiao Y, Wang M, Shen L, Shi Y, Feng S, Li C, Lin J, Tang S, Wu C. Development and validation of a scoring system to predict MASLD patients with significant hepatic fibrosis. Sci Rep 2025; 15:9639. [PMID: 40113920 PMCID: PMC11926222 DOI: 10.1038/s41598-025-91013-z] [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: 11/30/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
To address the need for a simple model to predict ≥ F2 fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD) patients, a study utilized data from 791 biopsy-proven MASLD patients from the NASH Clinical Research Network and Jinan University First Affiliated Hospital. The data were divided into training and internal testing sets through randomized stratified sampling. A multivariable logistic regression model using key categorical variables was developed to identify ≥ F2 fibrosis. External validation was performed using data from the FLINT trial and multiple centers in China. The DA-GAG score, incorporating diabetes, age, GGT, aspartate aminotransferase/ platelet ratio, and globulin/ total protein ratio, demonstrated superior performance in distinguishing ≥ F2 fibrosis with an area under the receiver operating characteristic curve of 0.79 in training and over 0.80 in testing datasets. The DA-GAG score efficiently identifies MASLD patients with ≥ F2 fibrosis, significantly reducing the medical burden.
Collapse
Affiliation(s)
- Linjing Long
- Department of Gastroenterology, the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, 510700, People's Republic of China
| | - Yue Wu
- Department of Hepatology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangdong, 510440, People's Republic of China
| | - Huijun Tang
- Department of Gastroenterology, Shenzhen Integrated Traditional Chinese and Western Medicine Hospital, Shenzhen, 518104, People's Republic of China
| | - Yanhua Xiao
- Department of Pathology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangdong, 510440, People's Republic of China
| | - Min Wang
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Lianli Shen
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Ying Shi
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Shufen Feng
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China
| | - Chujing Li
- Department of Hepatology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangdong, 510440, People's Republic of China
| | - Jiaheng Lin
- Department of Gastrointestinal Surgery, He Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, 510700, People's Republic of China
| | - Shaohui Tang
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China.
| | - Chutian Wu
- Department of Gastroenterology, the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, 510700, People's Republic of China.
- Department of Gastroenterology, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, People's Republic of China.
| |
Collapse
|
29
|
Zhang J, Zhou J, He Z, Xia Z, Liu H, Wu Y, Chen S, Wu B, Li H. Salidroside attenuates NASH through regulating bile acid-FXR/TGR5 signaling pathway via targeting gut microbiota. Int J Biol Macromol 2025; 307:142276. [PMID: 40118401 DOI: 10.1016/j.ijbiomac.2025.142276] [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: 01/10/2025] [Revised: 02/15/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Nonalcoholic steatohepatitis (NASH) is a significant threat to human health. Our previous study revealed that salidroside attenuated NASH and regulated the gut microbiota. However, whether the therapeutic effect of salidroside depends on gut microbiota remains to be determined. Therefore, we conducted further experiments to elucidate the essential functions of gut microbiota-associated metabolic pathways in the anti-NASH effects of salidroside. Our results showed that salidroside effectively alleviated lipid accumulation and inflammatory injury in NASH mice. 16S rRNA sequencing revealed that salidroside increased the abundance of Bacteroides. Mice receiving fecal microbiota transplantation (FMT) from salidroside-treated also presented less hepatic steatosis and higher abundance of Bacteroides. Antibiotics eliminated the effects of salidroside on hepatic steatosis and the gut microbiota. Mechanistically, salidroside and FMT from salidroside-treated altered the bile acid (BA) profile by decreasing the levels of conjugated BAs and tauro-α/β-muricholic acid and activated downstream farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5). Furthermore, we found that inhibitors of bile salt hydrolase (BSH) and FXR/TGR5 abolished the effects of salidroside and reduced downstream carnitine palmitoyltransferase 1α and lipoprotein lipase expression. These data demonstrate that salidroside attenuated NASH via gut microbiota-BA-FXR/TGR5 signaling pathway and reveal the underlying mechanism of salidroside on NASH.
Collapse
Affiliation(s)
- Jun Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Zheyun He
- Liver Diseases Institute, Ningbo No. 2 Hospital, Ningbo, Zhejiang 315000, China
| | - Zhanyang Xia
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Yuan Wu
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Si Chen
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China; Medical Experimental Department of Ningbo No.2 Hospital, Ningbo, Zhejiang 315000, China.
| |
Collapse
|
30
|
Lemmers SAM, Le Luyer M, Stoll SJ, Hoffnagle AG, Ferrell RJ, Gamble JA, Guatelli-Steinberg D, Gurian KN, McGrath K, O'Hara MC, Smith ADAC, Dunn EC. Inter-rater reliability of stress signatures in exfoliated primary dentition - Improving scientific rigor and reproducibility in histological data collection. PLoS One 2025; 20:e0318700. [PMID: 40106466 PMCID: PMC11922276 DOI: 10.1371/journal.pone.0318700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/20/2025] [Indexed: 03/22/2025] Open
Abstract
Accentuated Lines (ALs) in tooth enamel can reflect metabolic disruptions from physiological or psychological stresses during development. They can therefore serve as a retrospective biomarker of generalized stress exposure in archaeological and clinical research. However, little consensus exists on when ALs are identified and inter-rater reliability is poorly quantified across studies. Here, we sought to address this gap by examining the reliability of accentuated (AL) markings across raters, in terms of both the presence versus absence of ALs and their intensity (HAL= Highly Accentuated, MAL= Mildly Accentuated, RL= Retzius Line). Ratings were made and compared across observers (with different levels of experience) and pairs of raters (who agreed on AL coding through consensus meetings) (N = 15 teeth, eight observers). Results indicated that more experience in AL assessment does not necessarily produce higher reliability between raters. Most disagreements in intensity ratings occurred in categories other than HAL. Furthermore, when AL assessment was performed by pairs of raters, reliability was significantly higher than individual assessments (Gwet's AC1 = 0.28 to 0.56 for line presence assessment; Gwet's AC1 = 0.48 to 0.64 for line intensity assessment). Based on these results, we recommend a workflow called IRRISS (Improving Reliability and Reporting In Scoring of Stress-markers) to increase rigor and reproducibility in histological analysis of dental collections. The introduction of IRRISS is well-timed, given the surge in studies of teeth occurring across anthropological, epidemiological, medical, forensic, and climate research fields.
Collapse
Affiliation(s)
- Simone A M Lemmers
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States of America
- Elettra Sincrotrone Trieste S.C.p.A., Basovizza, Trieste, Italy
| | - Mona Le Luyer
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Samantha J Stoll
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Alison G Hoffnagle
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Rebecca J Ferrell
- National Science Foundation, Alexandria, Virginia, United States of America
| | - Julia A Gamble
- Department of Anthropology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Kaita N Gurian
- Department of Anthropology, The Ohio State University, Columbus, Ohio, United States of America
| | - Kate McGrath
- Department of Anthropology, SUNY Oneonta, New York, United States of America
- Center for the Advanced Study of Human Paleobiology, Department of Anthropology, The George Washington University, Washington District of Columbia, United States of America
- Centro Nacional de Investigación sobre la Evolución Humana, Burgos, Spain
| | - Mackie C O'Hara
- School of Anthropology and Conservation, University of Kent, Canterbury, United Kingdom
- Department of Sociology, College of Liberal Arts, Purdue University, West Lafayette, Indiana, United States of America
| | - Andrew D A C Smith
- Mathematics and Statistics Research Group, University of the West of England, Bristol, United Kingdom
| | - Erin C Dunn
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Sociology, College of Liberal Arts, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
31
|
Lai S, Tang D, Feng J. Mitochondrial targeted therapies in MAFLD. Biochem Biophys Res Commun 2025; 753:151498. [PMID: 39986088 DOI: 10.1016/j.bbrc.2025.151498] [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: 11/26/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a clinical-pathological syndrome primarily characterized by excessive accumulation of fat in hepatocytes, independent of alcohol consumption and other well-established hepatotoxic agents. Mitochondrial dysfunction is widely acknowledged as a pivotal factor in the pathogenesis of various diseases, including cardiovascular diseases, cancer, neurodegenerative disorders, and metabolic diseases such as obesity and obesity-associated MAFLD. Mitochondria are dynamic cellular organelles capable of modifying their functions and structures to accommodate the metabolic demands of cells. In the context of MAFLD, the excess production of reactive oxygen species induces oxidative stress, leading to mitochondrial dysfunction, which subsequently promotes metabolic disorders, fat accumulation, and the infiltration of inflammatory cells in liver and adipose tissue. This review aims to systematically analyze the role of mitochondria-targeted therapies in MAFLD, evaluate current therapeutic strategies, and explore future directions in this rapidly evolving field. We specifically focus on the molecular mechanisms underlying mitochondrial dysfunction, emerging therapeutic approaches, and their clinical implications. This is of significant importance for the development of new therapeutic approaches for these metabolic disorders.
Collapse
Affiliation(s)
- Sien Lai
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| | - Dongsheng Tang
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| | - Juan Feng
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| |
Collapse
|
32
|
El-Waseif AG, Elshal M, El-Kashef DH, Abu-Elsaad NM. Paricalcitol, an active vitamin D analog, mitigates dexamethasone-induced hepatic injury: Role of autophagy, pyroptosis, and PERK/Nrf2/HO-1 signaling pathway. Toxicol Appl Pharmacol 2025; 498:117307. [PMID: 40118256 DOI: 10.1016/j.taap.2025.117307] [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: 01/21/2025] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Drug-induced toxicity is considered a crucial clinical affair, as some adverse effects could be severe or life threatening. Drugs may have adverse effects by altering biological pathways that aren't always involved in the drug's reaction. From this perspective, the purpose of the current study was to assess the impacts of paricalcitol, a synthetic, active, and selective vitamin D receptor activator, on dexamethasone-induced liver injury, and discover the probable implicated signaling pathways as well. Male Wistar rats were treated with paricalcitol at a dose of 0.2 μg/kg, daily, i.p for 12 days and injected with 8 mg/kg dexamethasone i.p daily over the last 6 days. Administration of paricalcitol improved liver function markers, lipid profile, reduced histopathologic changes in hepatic sections, and restored normal oxidative status. Moreover, paricalcitol markedly decreased hepatic collagen deposition as confirmed by Masson's trichrome staining. Paricalcitol effectively inhibited endoplasmic reticulum stress through decreasing expression of tissue PERK and Chop, increasing hepatic Nrf2, and HO-1 activity. Besides, paricalcitol decreased levels of NLRP3 and IL-1β as well as decreased expression of active caspase-1 p20, GSDMD-N-terminal indicating suppression of NLRP3/caspase-1/GSDMD pyroptosis pathway. Paricalcitol can protect against dexamethasone-induced liver injury showing a promising therapeutic value in drug-induced liver injuries.
Collapse
Affiliation(s)
- Aamal G El-Waseif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt..
| | - Mahmoud Elshal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Nashwa M Abu-Elsaad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
33
|
He S, Lv Y, Qiu J, Cui S, Gao Z, Peng L. Ta 4C 3 MXene Slows Progression of Fatty Liver Disease through Its Anti-Inflammatory and ROS-Scavenging Effects. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17217-17229. [PMID: 40051029 DOI: 10.1021/acsami.4c20945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Treating metabolic dysfunction-associated fatty liver disease (MAFLD) and reducing the occurrence of MAFLD-associated liver cancer remain challenging. Two-dimensional (2D) tantalum carbide (Ta4C3) MXene nanozymes (MXenzymes) exhibit antioxidant and anti-inflammatory activities and have thus attracted considerable attention in the fields of oncology and engineering. However, the potential mechanism of action and bioactive properties of Ta4C3 in MAFLD remain uncertain. In our study, Ta4C3 not only inhibited lipid accumulation and disrupted lipid metabolism in hepatocytes but also reduced cell death caused by fatty acids by decreasing intracellular reactive oxygen species (ROS) levels, which significantly promoted the polarization of M1 macrophages to M2 macrophages by alleviating oxidative stress and further suppressing inflammatory factor expression. In mice fed a methionine-choline-deficient (MCD) diet, Ta4C3 reduced lipid accumulation, the infiltration of inflammatory cells, and liver cell apoptosis by modulating the cellular microenvironment through its anti-inflammatory and antioxidant properties. Therefore, Ta4C3 can be used as a multifunctional bioactive material to alleviate hepatic steatosis and inflammation in individuals with MAFLD/metabolic dysfunction-associated steatohepatitis (MASH) because of its robust antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Shuying He
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yuerong Lv
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Jingnan Qiu
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Shudan Cui
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Zixian Gao
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Liang Peng
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
- Department of Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| |
Collapse
|
34
|
Attanasi ML, Gregoski MJ, Rockey DC. Racial Differences in Liver Fibrosis Burden. Dig Dis Sci 2025:10.1007/s10620-025-08936-w. [PMID: 40102343 DOI: 10.1007/s10620-025-08936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/15/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND & AIMS Liver histology is the classic method for staging the severity of liver fibrosis, which in turn is an important predictor of clinical outcome. Here, we have hypothesized that the susceptibility to develop fibrosis varies among racial groups. METHODS We examined the histology of all patients over 18 years of age who underwent liver biopsy at the Medical University of South Carolina from 1/1/2013 to 7/1/2021. Patients with malignancy, liver metastases, or missing data were excluded. Fibrosis was quantified using the Batts-Ludwig system (F0 = no fibrosis to F4 = histological cirrhosis). Racial groups were propensity matched based on age, gender, diabetes, alcohol consumption, and CDC/ATSDR Social Vulnerability Index Themes to mitigate the risk of selection bias. RESULTS We identified 1101 patients with liver biopsy histological fibrosis scores. The cohort included 23% Black patients. After propensity matching, Black patients were significantly more likely to have Hepatitis C (73/228 (32%) vs 45/228 (20%), p < 0.001) and autoimmune hepatitis (34/228 (15%) vs 6/228 (3%), p < 0.001) than White patients, while White patients were significantly more likely to have metabolic dysfunction associated steatotic liver disease (71/228 (31%) vs 18/228 (8%), p < 0.001). White patients were significantly more likely to have cirrhosis than Black patients (White - 89/228 (39%) vs Black - 68/228 (30%), p < 0.05). CONCLUSION White patients had a greater overall burden of advanced fibrosis (F4/cirrhosis) than Black patients, independent of etiology. The data suggest that fibrosis risk and/or progression may be worse in White than Black patients.
Collapse
Affiliation(s)
- Michael L Attanasi
- Department of Internal Medicine, North Shore University Hospital, Manhasset, NY, USA
| | - Mathew J Gregoski
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, SC, USA
| | - Don C Rockey
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, SC, USA.
- Department of Internal Medicine, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 803, Charleston, SC, 29425, USA.
| |
Collapse
|
35
|
Kim HY, Lee KJ, Lee SS, Choi SJ, Kim DH, Heo S, Jang HJ, Choi SH. Diagnosis of moderate-to-severe hepatic steatosis using deep learning-based automated attenuation measurements on contrast-enhanced CT. Abdom Radiol (NY) 2025:10.1007/s00261-025-04872-5. [PMID: 40095018 DOI: 10.1007/s00261-025-04872-5] [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: 01/10/2025] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE To evaluate the utility of deep learning-based automated attenuation measurements on contrast-enhanced CT (CECT) for diagnosing moderate-to-severe hepatic steatosis (HS), using histology as reference standard. METHODS This retrospective study included 3,620 liver donors (2,393 men and 1,227 women; mean age, 31.7 ± 9.4 years), divided into the development (n = 2,714) and test (n = 906) cohorts. Attenuation values of the liver and spleen on CECT were measured both manually and using a deep learning algorithm (before and after radiologists' correction of segmentation errors). Performance of: (1) liver attenuation and (2) liver-spleen attenuation difference for diagnosing moderate-to-severe HS (> 33%) was assessed using the area under the receiver operating characteristic curve (AUC). Three different criteria targeting 95% sensitivity, 95% specificity, and the maximum Youden's index, respectively, for diagnosing moderate-to-severe HS, were developed and validated. RESULTS The performance of deep learning-based measurements did not differ significantly, with or without radiologists' corrections (p = 0.13). Liver-spleen attenuation difference outperformed liver attenuation alone in diagnosing moderate-to-severe HS in both deep learning-based (AUC, 0.868 vs. 0.821; p = 0.001) and manual (AUC, 0.871 vs. 0.823; p = 0.001) measurements. In the test cohort, the criterion targeting 95% sensitivity for diagnosing moderate-to-severe HS (liver-spleen attenuation difference ≤ 2.8 HU) yielded 92.0% (69/75) sensitivity and 48.5% (403/831) specificity. The criterion targeting 95% specificity (liver-spleen attenuation difference ≤ -18.8 HU) yielded 53.3% (40/75) sensitivity and 95.7% (795/831) specificity. The criterion targeting the maximum Youden's index (liver-spleen attenuation difference ≤ -8.2 HU) yielded 82.7% (62/75) sensitivity and 80.7% (671/831) specificity. CONCLUSION Deep learning-based automated measurements of liver and spleen attenuation on CECT can be used reliably to detect moderate-to-severe HS.
Collapse
Affiliation(s)
- Hae Young Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Jin Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Soo Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Se Jin Choi
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong Hwan Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Subin Heo
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyeon Ji Jang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Hyun Choi
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
36
|
Muscia Saez V, Perdicaro DJ, Cremonini E, Costantino VV, Fontana AR, Oteiza PI, Vazquez Prieto MA. Grape pomace extract attenuates high fat diet-induced endotoxemia and liver steatosis in mice. Food Funct 2025; 16:2515-2529. [PMID: 40029158 DOI: 10.1039/d4fo06332e] [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: 03/05/2025]
Abstract
Obesity is a prominent global health concern associated with chronic inflammation and metabolic disorders, such as insulin resistance, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD). Excessive consumption of saturated fats exacerbates these conditions by increasing intestinal barrier permeability and circulating endotoxins. This study aims to investigate, in a murine model of high-fat diet (HFD)-induced obesity, the potential beneficial effects of a grape pomace extract (GPE), rich in phenolic compounds, at mitigating endotoxemia, and liver steatosis. Underlying mechanisms were characterized in an in vitro model of intestinal inflammation and permeabilization, as induced by tumor necrosis factor alpha (TNFα) in Caco-2 cell monolayers. Consumption of a HFD (60% calories from fat) for 13 weeks induced obesity, insulin resistance, and liver damage, evidenced by higher levels of plasma alanine aminotransferase (ALT), hepatic triglycerides content, and steatosis. In addition, HFD caused metabolic endotoxemia, hepatic toll-like receptor 4 (TLR4) upregulation and inflammation. GPE supplementation significantly reduced body weight and subcutaneous and visceral adipose tissue weight, and attenuated metabolic dysregulation. Furthermore, GPE decreased circulating LPS levels and mitigated HFD-mediated hepatic TLR4 upregulation, nuclear factor kappa B (NF-κB) activation, and downstream expression of proteins involved in oxidative stress and inflammation (NOX4, TNFα, and F4/80). In Caco-2 cells, GPE mitigated TNFα-induced monolayer permeabilization, decreased tight junction (TJ) protein levels, enhanced cellular oxidant production, activated redox-sensitive signaling, i.e., NF-κB and ERK1/2, and increased NOX1 and MLCK mRNA levels, the latter being a key regulator of monolayer permeability. The above findings suggest that GPE may protect against HFD-induced obesity and associated metabolic dysfunction (insulin resistance and NAFLD) by modulating intestinal barrier integrity and related endotoxemia.
Collapse
Affiliation(s)
- V Muscia Saez
- Laboratorio de Nutrición y Fisiopatología de la Obesidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, M5502JMA, Mendoza, Argentina.
| | - D J Perdicaro
- Laboratorio de Nutrición y Fisiopatología de la Obesidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, M5502JMA, Mendoza, Argentina.
| | - E Cremonini
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, USA
| | - V V Costantino
- Laboratorio de Fisiopatología Renal, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, Argentina
| | - A R Fontana
- Laboratorio de Bioquímica Vegetal, Instituto de Biología Agrícola de Mendoza (IBAM), Facultad de Ciencias Agrarias, CONICET-Universidad Nacional de Cuyo, M5528AHB, Chacras de Coria, Argentina
| | - P I Oteiza
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, USA
| | - M A Vazquez Prieto
- Laboratorio de Nutrición y Fisiopatología de la Obesidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, M5502JMA, Mendoza, Argentina.
| |
Collapse
|
37
|
Brush M, Auh S, Cochran E, Tuska R, Koh C, Kleiner DE, Lightbourne M, Brown RJ. Effects of Metreleptin in Patients With Generalized Lipodystrophy Before vs After the Onset of Severe Metabolic Disease. J Clin Endocrinol Metab 2025; 110:e1051-e1061. [PMID: 38757950 PMCID: PMC11913101 DOI: 10.1210/clinem/dgae335] [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: 02/22/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT Leptin replacement therapy with metreleptin improves metabolic abnormalities in patients with generalized lipodystrophy (GLD). OBJECTIVE Determine how timing of metreleptin initiation in the clinical course of GLD affects long-term metabolic health. METHODS Retrospective analysis of patients ≥6 months old with congenital (n = 47) or acquired (n = 16) GLD treated with metreleptin at the National Institutes of Health since 2001. Least squares means for glycated hemoglobin (HbA1c), insulin area under the curve from oral glucose tolerance tests, triglycerides, urine protein excretion, platelets, transaminases, and aspartate aminotransferase (AST) to Platelet Ratio Index for early and late treatment groups, defined by baseline metabolic health, were analyzed during median 72 (24-108) months' follow-up. RESULTS Compared to late groups, early groups based on metabolic status had higher mean ± SEM insulin area under the curve (20 831 ± 1 vs 11 948 ± 1), lower HbA1c (5.3 ± 0.3 vs 6.8 ± 0.3%), triglycerides (101 ± 1 vs 193 ± 1 mg/dL), urine protein excretion (85 ± 1.5 vs 404 ± 1.4 mg/24 h), alanine aminotransferase (30 ± 1 vs 53 ± 1 U/L), AST (23 ± 1 vs 40 ± 1 U/L), and AST to Platelet Ratio Index (0.22 ± 1.3 vs 0.78 ± 1.3), and higher platelets (257 ± 24 vs 152 ± 28 K/µL) during follow-up (P < .05). Compared to patients ≥6 years old at baseline, patients <6 years had lower HbA1c (4.5 ± 0.5 vs 6.4 ± 0.2%) and higher AST (40 ± 1vs 23 ± 1 U/L) during follow (P < .05). CONCLUSION Patients with GLD who initiated metreleptin before the onset of severe metabolic complications had better long-term control of diabetes, proteinuria, and hypertriglyceridemia. Early treatment may also result is less severe progression of liver fibrosis, but further histological studies are needed to determine the effects of metreleptin therapy on liver disease.
Collapse
Affiliation(s)
- Maiah Brush
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sungyoung Auh
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elaine Cochran
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca Tuska
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Koh
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David E Kleiner
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marissa Lightbourne
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
38
|
Hara E, Ohshima K, Takimoto M, Bai Y, Hirata M, Zeng W, Uomoto S, Todoroki M, Kobayashi M, Kozono T, Kigata T, Shibutani M, Yoshida T. Flutamide Promotes Early Hepatocarcinogenesis Through Mitophagy in High-Fat Diet-Fed Non-Obese Steatotic Rats. Int J Mol Sci 2025; 26:2709. [PMID: 40141351 PMCID: PMC11943065 DOI: 10.3390/ijms26062709] [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: 12/22/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Flutamide (FL), a non-steroidal drug used for its antiandrogenic, anticancer, and disrupting endocrine properties, induces mitochondrial toxicity and drug metabolism enzymes and promotes hepatocarcinogenesis. The inhibition of mitophagy, leading to the accumulation of damaged mitochondria, is implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). In this study, we investigated the effects of FL in high-fat diet (HFD)-induced non-obese steatosis rats, categorized into four groups: basal diet (BD), BD + FL, HFD, and HFD + FL. The FL exacerbated HFD-induced steatosis and marginally increased preneoplastic lesions. To analyze hepatic preneoplastic lesions, we divided them into clusters based on the expression ratios of the mitophagy regulators LC3 and AMBRA1. The expression rates of LC3 and AMBRA1 in these precancerous lesions were classified into three clusters using k-means clustering. The HFD group exhibited an increased ratio of mitophagy inhibition clusters, as indicated by decreased LC3 and increased AMBRA1 levels in background hepatocytes and preneoplastic lesions. FL counteracted HFD-mediated mitophagy inhibition, as indicated by increased LC3 and decreased AMBRA1 levels in background hepatocytes. Our clustering analysis revealed that FL-induced mitophagy induction relied on Parkin expression. The present study underscores the significance of cluster analysis in understanding the role of mitophagy within small preneoplastic lesions and suggests that FL may potentially exacerbate NAFLD-associated hepatocarcinogenesis by affecting mitophagy.
Collapse
Affiliation(s)
- Emika Hara
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Kanami Ohshima
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Mio Takimoto
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Yidan Bai
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Mai Hirata
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Wen Zeng
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Suzuka Uomoto
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Mai Todoroki
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
- Cooperative Division of Veterinary Sciences, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
- Cooperative Division of Veterinary Sciences, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Takuma Kozono
- Smart-Core-Facility Promotion Organization, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan;
| | - Tetsuhito Kigata
- Laboratory of Veterinary Anatomy, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan;
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Cooperative Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan (M.S.)
| |
Collapse
|
39
|
Gomes SP, de Castro GS, de Oliveira VPS, Cogliati B, Iacopino AGDS, Pires ISDO, Schimming BC, Dias FGG, Kfoury JR, Sasahara THDC. Hepatic stereological analysis in obese Zucker rats (Leprfa) with dyslipidemia. Acta Cir Bras 2025; 40:e402325. [PMID: 40105603 PMCID: PMC11908736 DOI: 10.1590/acb402325] [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: 08/01/2024] [Accepted: 01/07/2025] [Indexed: 03/20/2025] Open
Abstract
PURPOSE To characterize histologically and stereologically the hepatic steatosis in obese Zucker (fat, n = 6, with a mutation in the leptin receptor - Leprfa) and control Zucker (lean, n = 6) rats, analyzing macroscopic and microscopic differences to understand the influence of obesity on hepatic pathology. METHODS Zucker rats were fed standard chow for 90 days. Macroscopic, qualitative, and histoquantitative (stereological) approaches were used, involving body and liver weight measurement, morphological analysis, and histopathological classification of metabolic dysfunction-associated steatotic liver disease. RESULTS Zucker fat rats had higher body weight (p = 0.0022), liver weight (p = 0.0022), serum total cholesterol (p = 0.0022), and triacylglycerol (p = 0.0022) compared to Zucker lean rats. Stereological analysis showed that hepatocyte volume density (p = 0.0022) and total hepatocyte volume (p = 0.0001) were lower, and the volume density (p = 0.002) and total volume of steatosis (p = 0.002) were higher in Zucker fat rats compared to lean rats. CONCLUSION The findings indicated that obesity induces significant alterations in the hepatic morphology of Zucker rats, showing that hepatocyte volume is lower in obese animals. This study reinforces the utility of the obese Zucker rat model to investigate the effects of obesity on liver health and suggests hepatic steatosis requires therapeutic strategies focused on modulating these parameters.
Collapse
Affiliation(s)
- Silvio Pires Gomes
- Universidade de São Paulo – Faculdade de Medicina Veterinária e Zootecnia – Departamento de Cirurgia – São Paulo (SP) – Brazil
- Universidade de São Paulo – Faculdade de Medicina – Departamento de Cirurgia – São Paulo (SP) – Brazil
| | - Gabriela Salim de Castro
- Universidade de São Paulo – Faculdade de Medicina – Departamento de Cirurgia – São Paulo (SP) – Brazil
| | - Vinicius Pedro Silva de Oliveira
- Universidade de São Paulo – Faculdade de Medicina Veterinária e Zootecnia – Departamento de Cirurgia – São Paulo (SP) – Brazil
- Centro Universitário Braz Cubas – Faculdade de Medicina Veterinária – Mogi das Cruzes (SP) – Brazil
| | - Bruno Cogliati
- Universidade de São Paulo – Faculdade de Medicina Veterinária e Zootecnia – Departamento de Patologia – São Paulo (SP) – Brazil
| | - Andressa Galvão da Silva Iacopino
- Universidade de São Paulo – Faculdade de Medicina Veterinária e Zootecnia – Departamento de Cirurgia – São Paulo (SP) – Brazil
- Universidade Nove de Julho – Faculdade de Medicina Veterinária – São Paulo (SP) – Brazil
| | | | - Bruno Cesar Schimming
- Universidade Estadual Paulista – Instituto de Biociências – Departamento de Biologia Estrutural e Funcional – Botucatu (SP) – Brazil
| | | | - José Roberto Kfoury
- Universidade de São Paulo – Faculdade de Medicina Veterinária e Zootecnia – Departamento de Cirurgia – São Paulo (SP) – Brazil
| | - Tais Harumi de Castro Sasahara
- Universidade de São Paulo – Faculdade de Medicina Veterinária e Zootecnia – Departamento de Cirurgia – São Paulo (SP) – Brazil
- Universidade Estadual Paulista – Instituto de Biociências – Departamento de Biologia Estrutural e Funcional – Botucatu (SP) – Brazil
| |
Collapse
|
40
|
Arai J, Okumura A, Kimoto S, Sakamoto K, Kitada T, Kitano R, Inoue T, Nishimura S, Inden N, Muraki Y, Kato N, Ito K. Efficacy of measuring natural killer-activating receptor ligands to predict the pathogenesis of metabolic dysfunction-associated steatotic liver disease. Hepatol Int 2025:10.1007/s12072-025-10800-y. [PMID: 40085416 DOI: 10.1007/s12072-025-10800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/15/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE The proportion of non-B/non-C hepatocellular carcinoma cases is increasing, and the principal cause is metabolic dysfunction-associated steatotic liver disease (MASLD). The degree of intrahepatic natural killer (NK) cell infiltration has been reported to correlate with MASLD progression. However, reports on MASLD are limited. We aimed to investigate the involvement of NK cell-activating receptor ligands in MASLD pathogenesis. METHODS This study cohort comprised 69 patients with biopsy-proven MASLD treated between 2012 and 2018 at our institute. The concentrations of major histocompatibility complex class I polypeptide-related sequences A and B (MICA and MICB, respectively) and B7H6 in patient sera were measured using enzyme-linked immunosorbent assay kits. Data were statistically compared between those with metabolic-associated steatotic liver (MASL, n = 25) and those with metabolic dysfunction-associated steatohepatitis (MASH, n = 44). The clinical characteristics related to higher concentrations of each NK cell-activating receptor ligand were also investigated. RESULTS The MASH group had a higher level of the ligands than the MASL group. Furthermore, the MASH group had a significantly higher level of the Mac-2-binding protein glycosylation isomer (M2BPGi) than the MASL group (p < 0.001). MICA and MICB were positively correlated, and all three ligands were strongly correlated with alpha-fetoprotein and protein induced by vitamin K absence 2. Although MICB levels positively correlated with aspartate transaminase and alanine transaminase levels (p < 0.005), patients with higher MICA and B7H6 levels had higher M2BPGi levels. Interestingly, concentrations of B7H6 were significantly correlated with portal inflammation (p < 0.001), rather than lobular inflammation. CONCLUSION The three NK-activating receptor ligands were higher in the sera of the MASH group than those of the MASL group and strongly correlated with tumor markers, indicating the potential for hepatocarcinogenesis. Higher concentrations of serum B7H6 were correlated with advanced fibrosis and the degree of portal inflammation, which is a potential biomarker for predicting the pathogenesis of MASH.
Collapse
Affiliation(s)
- Jun Arai
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| | - Akinori Okumura
- Department of Microbiology and Immunology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Satoshi Kimoto
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Kazumasa Sakamoto
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Tomoya Kitada
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Rena Kitano
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Tadahisa Inoue
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Sayaka Nishimura
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Noriko Inden
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Yukiko Muraki
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kiyoaki Ito
- Department of Gastroenterology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| |
Collapse
|
41
|
Chen L, Li Y, Patel RN, Sottas C, Raul MC, Patel ND, Zambidis A, Li M, Chopra S, Papadopoulos V. AAV8-mediated silencing of Atad3 prevents the progression from simple steatosis to MASH in mice by reduced IL6 secretion. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167792. [PMID: 40086517 DOI: 10.1016/j.bbadis.2025.167792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/19/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
ATAD3A deficiency in hepatocytes has been shown to promote simple steatosis (SS), ATAD3 is upregulated in MCD diet-induced MASH. Since the MCD diet is commonly used to induce liver fibrosis, which is related to HSCs activation, we are prompted to investigate the functions of ATAD3 in these two cell types and their mediated transition from SS to MASH. To investigate the role of ATAD3A in HSCs, human LX-2 cells were treated with TGFβ. The results showed that ATAD3A expression was linked to the fibrotic markers ACTA2 and COL1A1. Knockdown of ATAD3A reversed TGFβ-induced HSC activation by downregulating both canonical (SMAD2/3) and non-canonical (ERK1/2 and p38 MAPK) TGFβ signaling pathways. To examine the effect of ATAD3 on the transition from SS to MASH, MASH was induced in mice using the GAN diet for 24 weeks. After 12 weeks, AAV8-conjugated Atad3 shRNA was administered to knock down Atad3 in the liver. This intervention suppressed steatosis and fibrosis while enhancing insulin sensitivity. Further analysis using conditioned medium (CM) from WT and ATAD3A KO Huh7 cells treated with LPS and PA revealed that IL-6 secretion from Huh7 hepatocytes activated HSCs. However, IL-6 secretion was diminished in ATAD3A KO CM. CM from ATAD3A KO cells also suppressed expression of fibrotic markers ACTA2, PP38, and P-SMAD3 compared to WT cells under MASH conditions. These data suggest that AAV8-mediated Atad3 silencing in hepatocytes prevents the transition from SS to MASH, at least in part, by downregulating IL-6 secretion to suppress HSC activation in MASH.
Collapse
Affiliation(s)
- Liting Chen
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuchang Li
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Rahil Nitinkumar Patel
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Chantal Sottas
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Mahima Chandrakant Raul
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Nrupa Dinesh Patel
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Alexander Zambidis
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Meng Li
- USC Libraries Bioinformatic Services of the University of Southern California, Los Angeles, CA 90033, USA
| | - Shefali Chopra
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
42
|
Luo X, Deng H, Li Q, Zhao M, Zhang Y, Guo J, Wen Y, Chen G, Li J. Bulk transcriptome and single-nucleus RNA sequencing analyses highlight the role of recombination activating 1 in non-alcoholic fatty liver disease. Int J Biol Macromol 2025; 307:141919. [PMID: 40074128 DOI: 10.1016/j.ijbiomac.2025.141919] [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: 10/13/2024] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic condition with an incompletely understood pathogenesis. In this study, five candidate genes-RAG1, CKAP2, CENPK, TYMS, and BUB1-were identified as being associated with NAFLD progression through integrative bioinformatics analyses. A predictive model incorporating these genes demonstrated strong robustness and diagnostic accuracy. Single-nucleus RNA sequencing analysis further revealed that RAG1 plays a potential role in hepatocytes of NAFLD patients. Functional experiments using RNA interference to suppress RAG1 expression in HepG2 cells treated with oleic and palmitic acids showed reduced total glyceride and cholesterol levels, mitigated lipid accumulation, and alterations in pathways related to lipid metabolism, inflammation, and fibrosis. Furthermore, adeno-associated virus-specific knockdown of RAG1 in hepatocytes attenuated hepatic steatosis in high-fat diet-fed mice. These findings suggest that investigating the molecular mechanisms of hub genes like RAG1 may advance our understanding of NAFLD pathogenesis and inform therapeutic development.
Collapse
Affiliation(s)
- Xiaohua Luo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Hongbo Deng
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Qiang Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Miao Zhao
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Science, Central South University, 410078 Changsha, China
| | - Yu Zhang
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junjie Guo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yifan Wen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Guangshun Chen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| | - Jiequn Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
43
|
Brzozowska N, Wu LYD, Khodzhaeva V, Griffiths WJ, Duckworth A, Jung H, Coorens THH, Hooks Y, Chambers JE, Campbell PJ, Marciniak SJ, Hoare M. Selection for somatic escape variants in SERPINA1 in the liver of patients with alpha-1 antitrypsin deficiency. Nat Genet 2025:10.1038/s41588-025-02125-1. [PMID: 40065168 DOI: 10.1038/s41588-025-02125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
Somatic variants accumulate in non-malignant tissues with age. Functional variants, leading to clonal advantage of hepatocytes, accumulate in the liver of patients with acquired chronic liver disease (CLD). Whether somatic variants are common to CLD from differing etiologies is unknown. We analyzed liver somatic variants in patients with genetic CLD from alpha-1 antitrypsin (A1AT) deficiency or hemochromatosis. We show that somatic variants in SERPINA1, the gene encoding A1AT, are strongly selected for in A1AT deficiency, with evidence of convergent evolution. Acquired SERPINA1 variants are clustered at the carboxyl terminus of A1AT, leading to truncation. In vitro and in vivo, C-terminal truncation variants reduce disease-associated Z-A1AT polymer accumulation and disruption of the endoplasmic reticulum, supporting the C-terminal domain swap mechanism. Therefore, somatic escape variants from a deleterious germline variant are selected for in A1AT deficiency, suggesting that functional somatic variants are disease-specific in CLD and point to disease-associated mechanisms.
Collapse
Affiliation(s)
| | - Lily Y D Wu
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Vera Khodzhaeva
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Adam Duckworth
- Department of Pathology, Addenbrooke's Hospital, Cambridge, UK
| | | | - Tim H H Coorens
- Wellcome Trust Sanger Institute, Hinxton, UK
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Joseph E Chambers
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | | | - Stefan J Marciniak
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Matthew Hoare
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Early Cancer Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
44
|
Huang DQ, Wong VWS, Rinella ME, Boursier J, Lazarus JV, Yki-Järvinen H, Loomba R. Metabolic dysfunction-associated steatotic liver disease in adults. Nat Rev Dis Primers 2025; 11:14. [PMID: 40050362 DOI: 10.1038/s41572-025-00599-1] [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: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the umbrella term that comprises metabolic dysfunction-associated steatotic liver, or isolated hepatic steatosis, through to metabolic dysfunction-associated steatohepatitis, the progressive necroinflammatory disease form that can progress to fibrosis, cirrhosis and hepatocellular carcinoma. MASLD is estimated to affect more than one-third of adults worldwide. MASLD is closely associated with insulin resistance, obesity, gut microbial dysbiosis and genetic risk factors. The obesity epidemic and the growing prevalence of type 2 diabetes mellitus greatly contribute to the increasing burden of MASLD. The treatment and prevention of major metabolic comorbidities such as type 2 diabetes mellitus and obesity will probably slow the growth of MASLD. In 2023, the field decided on a new nomenclature and agreed on a set of research and action priorities, and in 2024, the US FDA approved the first drug, resmetirom, for the treatment of non-cirrhotic metabolic dysfunction-associated steatohepatitis with moderate to advanced fibrosis. Reliable, validated biomarkers that can replace histology for patient selection and primary end points in MASH trials will greatly accelerate the drug development process. Additionally, noninvasive tests that can reliably determine treatment response or predict response to therapy are warranted. Sustained efforts are required to combat the burden of MASLD by tackling metabolic risk factors, improving risk stratification and linkage to care, and increasing access to therapeutic agents and non-pharmaceutical interventions.
Collapse
Affiliation(s)
- Daniel Q Huang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore
| | - Vincent W S Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Mary E Rinella
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Jerome Boursier
- Service d'Hépato-Gastroentérologie et Oncologie Digestive, Centre Hospitalier Universitaire d'Angers, Angers, France
- Laboratoire HIFIH, SFR ICAT 4208, Université d'Angers, Angers, France
| | - Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, San Diego, CA, USA.
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California at San Diego, San Diego, CA, USA.
| |
Collapse
|
45
|
Ou-Yang K, He Y, Yang H, Wang L, Zhang Q, Li D, Li L. Microcystin-LR induces fatty liver metabolic disease in zebrafish through the PPARα-NOD1 pathway: In vivo, in vitro, and in silico investigations. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136813. [PMID: 39657491 DOI: 10.1016/j.jhazmat.2024.136813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Hepatic lipid metabolism dysfunction caused by cyanobacteria bloom-released microcystin-LR (MC-LR) contributes to the development of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis (NASH), thereby severely impacting the health and safety of animals and humans. In this study, the effects and mechanisms of different environmental concentrations of MC-LR (0, 0.1, 1, and 10 μg/L) on fatty liver metabolic disease in zebrafish were investigated using in vivo, in vitro, and in silico models. Exposure to 10 μg/L of MC-LR-induced NASH in zebrafish, characterized by hepatic steatosis, toxic saturated fatty acid (SFA) accumulation, and inflammation. Analyses of the liver transcriptome, molecular docking, molecular dynamics simulation, and in vitro experiments indicated that PPARα might be a key molecular target in MC-LR-induced steatosis and in toxic-SFA accumulation. The results obtained from molecular docking, molecular dynamics simulation, and NOD1-inhibitor experiments further revealed that MC-LR-derived SFAs, such as palmitic acid, could target the NOD1 protein to initiate hepatitis in zebrafish. The benchmark dose model identified palmitic acid as a sensitive indicator of MC-LR-induced NASH, and the point of departure value was estimated to be 1.634 μg/L. In conclusion, our findings offer new insights into the mechanism of MC-LR-induced NASH and aid in the prognosis and treatment of MC-LR-related liver metabolic diseases, as well as in assessing the health risks associated with cyanobacterial blooms.
Collapse
Affiliation(s)
- Kang Ou-Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ya He
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Hui Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Liangmou Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Qian Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Dapeng Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, PR China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, PR China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, PR China
| | - Li Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, PR China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, PR China; Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, PR China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, PR China.
| |
Collapse
|
46
|
Bertran L, Capellades J, Abelló S, Richart C. Untargeted lipidomic analysis of metabolic dysfunction-associated steatohepatitis in women with morbid obesity. PLoS One 2025; 20:e0318557. [PMID: 40036208 DOI: 10.1371/journal.pone.0318557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/19/2025] [Indexed: 03/06/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatohepatitis (MASH) represents the severe condition of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Currently, there is a need to identify non-invasive biomarkers for an accurate diagnosis of MASH. Previously, omics studies identified alterations in lipid metabolites involved in MASLD. However, these studies require validation in other cohorts. In this sense, our aim was to perform lipidomics to identify the circulating lipid metabolite profile of MASH. We assessed a liquid chromatography coupled to a mass spectrometer-based untargeted lipidomic assay in serum samples of 216 women with morbid obesity that were stratified according to their hepatic diagnosis into Normal Liver (NL, n = 44), Simple Steatosis (SS, n = 66) and MASH (n = 106). First, we identified a profile of lipid metabolites that are increased in MASLD, composed of ceramides, triacylglycerols (TAG) and some phospholipids. Then, we identified that patients with SS have a characteristic profile of increased levels of ceramides, diacylglycerols DG (36:2) and DG (36:4), some TAG and a few phospholipids such as PC (32:1), PE (38:3), PE (40:6), PI (32:0) and PI (32:1). Later, in MASH patients, we found increased levels of ceramides, deoxycholic acid, a set of TAG, and some phospholipids such as PC, PE, PI and LPI; while we found decreased levels of the DG (36:0). Finally, we have reported a panel of lipid metabolites that might be used to differentiate patients with MASH from SS patients, made up of increased levels of 9-HODE some PC and PE, the LPI (16:0) and decreased levels of DG (36:0). To conclude, our investigation has suggested a lipid metabolite profile associated with MASLD and MASH. Specifically, a set of lipid metabolites seems to be discriminatory in MASH subjects compared to SS individuals. Thus, this panel of lipid metabolites could be used as a non-invasive diagnostic tool.
Collapse
Affiliation(s)
- Laia Bertran
- Department of Medicine and Surgery, Rovira i Virgili University, Tarragona, Spain
| | - Jordi Capellades
- Department of Electronic, Electric and Automatic Engineering, Higher Technical School of Engineering, Rovira i Virgili University, Tarragona, Spain
| | - Sonia Abelló
- Scientific and Technical Service, Rovira i Virgili University, Tarragona, Spain
| | - Cristóbal Richart
- Department of Medicine and Surgery, Rovira i Virgili University, Tarragona, Spain
| |
Collapse
|
47
|
Sheikh MY, Hasan N, Almozuaghi M, Akhtar NM, Grewal Y, Schneider C. Accuracy of Velacur in Assessing MASLD and MASH Patients Using Biopsy as the Gold Standard. Diagnostics (Basel) 2025; 15:615. [PMID: 40075862 PMCID: PMC11898527 DOI: 10.3390/diagnostics15050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: VelacurTM is a novel, point-of-care ultrasound device developed to accurately diagnose patients with Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Metabolic Dysfunction-Associated Steatohepatitis (MASH). The Velacur system non-invasively assesses liver stiffness, attenuation, and the Velacurdetermined fat fraction (VDFF). In this study, the performance of Velacur was measured against biopsy results in a cohort of MASLD and MASH patients. Methods: This prospective study enrolled adult patients who were scheduled to undergo biopsy within 6 months of enrollment. The primary objective was to validate Velacur's findings against that of histological findings. The secondary objective was to compare Velacur results with those of FibroScan. Results: A total of 78 participants were enrolled, and 70 were included in the analysis. Patients had a mean age of 53.3 ± 13.1 years, with a mean BMI of 35.0 ± 6.24 kg/m2. A total of 11, 19, 13, 25, and 2 were characterized as F0 to F4, respectively. The mean Velacur stiffness was 6.48 ± 1.4 kPa, and the mean VDFF was 14.4 ± 5.1%. In patients with significant fibrosis the Velacur AUC [95% CI] was 0.86 [0.76, 0.93] and 0.79 [0.66, 0.88] for patients with advanced fibrosis. For measurements of steatosis, 2, 24, 20, and 24 patients were found to have S0 to S3, respectively. To determine moderate steatosis (≥S2), the VDFF had an AUC of 0.846 [0.716, 0.920]. In the comparison population (n = 59), VDFF (0.85 [0.72, 0.94]) was significantly different than FibroScan CAP (0.50 [0.35, 0.66]) for the detection of moderate steatosis. Conclusions: This study validates the use of Velacur as a non-invasive tool for assessment of steatosis and fibrosis, hallmarks of MASLD and MASH, when compared to histological evidence provided via hepatic biopsy. Further, Velacur outperformed FibroScan in the assessment of steatosis.
Collapse
Affiliation(s)
- Muhammad Y. Sheikh
- Fresno Clinical Research Center, Fresno, CA 93720, USA; (M.Y.S.); (N.H.); (M.A.); (N.M.A.); (Y.G.)
| | - Nameer Hasan
- Fresno Clinical Research Center, Fresno, CA 93720, USA; (M.Y.S.); (N.H.); (M.A.); (N.M.A.); (Y.G.)
| | - Marwan Almozuaghi
- Fresno Clinical Research Center, Fresno, CA 93720, USA; (M.Y.S.); (N.H.); (M.A.); (N.M.A.); (Y.G.)
| | - Nadeem M. Akhtar
- Fresno Clinical Research Center, Fresno, CA 93720, USA; (M.Y.S.); (N.H.); (M.A.); (N.M.A.); (Y.G.)
| | - Yugjeet Grewal
- Fresno Clinical Research Center, Fresno, CA 93720, USA; (M.Y.S.); (N.H.); (M.A.); (N.M.A.); (Y.G.)
| | | |
Collapse
|
48
|
Baumert B, Maretti-Mira A, Walker D, Li Z, Stratakis N, Wang H, Zhao Y, Fischer F, Jia Q, Valvi D, Bartell S, Chen J, Inge T, Ryder J, Jenkins T, Sisley S, Xanthakos S, Kleiner D, Kohli R, Rock S, Eckel S, Merrill ML, Aung M, Salomon M, McConnell R, Goodrich J, Conti D, Golden-Mason L, Chatzi L. Integrated Spheroid-to-Population Framework for Evaluating PFHpA-Associated Metabolic Dysfunction and Steatotic Liver Disease. RESEARCH SQUARE 2025:rs.3.rs-5960979. [PMID: 40092438 PMCID: PMC11908348 DOI: 10.21203/rs.3.rs-5960979/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The rising prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), particularly among pediatric populations, requires identification of modifiable risk factors to control disease progression. Per- and polyfluoroalkyl substances (PFAS) have emerged as potential contributors to liver damage; however, their role in the etiology of MASLD remains underexplored. This study aimed to bridge the gap between human epidemiological data and in vitro experimental findings to elucidate the effect of perfluoroheptanoic acid (PFHpA), a short chain, unregulated PFAS congener on MASLD development. Our analysis of the Teen-LABS cohort, a national multi-site study on obese adolescents undergoing bariatric surgery, revealed that doubling of PFHpA plasma levels was associated with an 80% increase in MASLD risk (OR, 1.8; 95% CI: 1.3-2.5) based on liver biospies. To further investigate the underlying mechanisms, we used 3D human liver spheroids and single-cell transcriptomics to assess the effect of PFHpA on hepatic metabolism. Integrative analysis identified dysregulation of common pathways in both human and spheroid models, particularly those involved in innate immunity, inflammation, and lipid metabolism. We applied the latent unknown clustering with integrated data (LUCID) model to assess associations between PFHpA exposure, multiomic signatures, and MASLD risk. Our results identified a proteome profile with significantly higher odds of MASLD (OR = 7.1), whereas a distinct metabolome profile was associated with lower odds (OR = 0.51), highlighting the critical role of protein dysregulation in disease pathogenesis. A translational framework was applied to uncover the molecular mechanisms of PFAS-induced MASLD in a cohort of obese adolescents. Identifying key molecular mechanisms for PFAS-induced MASLD can guide the development of targeted prevention and treatment.
Collapse
|
49
|
Miao GL, Zhang WX, Xu YT, Liu YR, Lai PP, Guo JB, Chen GL, Chen JX, Zhou ZH, Li YW, Zhang C, Ding Y, Zhang LX, Han YF, Chen JX, Wu JD, Zhao YQ, Mei S, Zhao Y, Ma YW, Zhang L, Huang W, Zhao DY, Dong ED, Wang YH, Xian XD. Motor protein KIF13B orchestrates hepatic metabolism to prevent metabolic dysfunction-associated fatty liver disease. Mil Med Res 2025; 12:11. [PMID: 40038775 DOI: 10.1186/s40779-025-00594-3] [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: 08/16/2024] [Accepted: 01/16/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Kinesin family member 13B (KIF13B), a crucial motor protein, exerts multiple cellular biological functions. However, the implication of KIF13B in metabolic dysfunction-associated fatty liver disease (MAFLD) has not been explored yet. This study aimed to investigate KIF13B's role and underlying mechanism in MAFLD and proposes it as a potential pharmacological target. METHODS We assessed KIF13B expression in MAFLD patients and rodent models. The roles of Kif13b in lipid metabolism and MAFLD were investigated using whole-body Kif13b knockout mice, hepatocyte-specific Kif13b-deficient mice and hamsters exposed to different diets. The underlying mechanisms by which Kif13b governed hepatic lipid homeostasis and MAFLD progression were explored in vitro. Finally, the Kif13b's impact on atherosclerotic development was studied in the context of MAFLD. RESULTS KIF13B expression was reduced in patients and murine models with MAFLD. Rodents with global or liver-specific knockout of the Kif13b gene exhibit spontaneous hepatic steatosis, which is further exacerbated by different overnutrition diets. Overexpression of human KIF13B by lentivirus effectively prevented metabolic dysfunction-associated steatohepatitis (MASH) in methionine-choline-deficient diet (MCD)-fed mice. Furthermore, Kif13b deficiency accelerates atherosclerosis in the context of MAFLD. Mechanistically, Kif13b depletion increases hepatic lipid synthesis and impairs mitochondrial oxidative phosphorylation. Further screening reveals that Kif13b interacts with AMP-activated catalytic subunit alpha 1 (AMPKα1) to regulate the phosphorylation of AMPKα1, governing mitochondrial homeostasis and suppressing sterol regulatory element binding protein 1 (Srebp1)-mediated de novo lipogenesis in the liver. CONCLUSION This work establishes a causal relationship between KIF13B deficiency and MAFLD, emphasizing KIF13B as a potential therapeutic target for treating MAFLD.
Collapse
Affiliation(s)
- Guo-Lin Miao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Wen-Xi Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yi-Tong Xu
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yi-Ran Liu
- Department of Biomedical Informatics, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ping-Ping Lai
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jia-Bao Guo
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Gong-Lie Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jing-Xuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Zi-Hao Zhou
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yan-Wei Li
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, 110134, China
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, 110134, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, 110134, China
| | - Lian-Xin Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yu-Fei Han
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jin-Xuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jing-Dong Wu
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education, Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Yin-Qi Zhao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Si Mei
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education, Key Laboratory of Cell Proliferation and Differentiation, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Center for Life Sciences, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Yuan-Wu Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, 100021, China
| | - Ling Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Wei Huang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Dong-Yu Zhao
- Department of Biomedical Informatics, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Er-Dan Dong
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266113, Shandong, China.
| | - Yu-Hui Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Xun-De Xian
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
50
|
Dong L, Lou W, Xu C, Wang J. Naringenin cationic lipid-modified nanoparticles mitigate MASLD progression by modulating lipid homeostasis and gut microbiota. J Nanobiotechnology 2025; 23:168. [PMID: 40038718 DOI: 10.1186/s12951-025-03228-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
Naringenin (NAR) possesses various pharmacological activities including antioxidant, anti-inflammatory, and hepatoprotective effects. However, its therapeutic efficacy is limited by its hydrophobic and crystalline nature. This study aimed to investigate the therapeutic potential and molecular mechanisms of NAR efficiently loaded into cationic nanoparticles (NP-NAR) for treating metabolic dysfunction-associated steatotic liver disease (MASLD) in a mouse model. The results demonstrated that NP-NAR effectively ameliorated lipid metabolism dysbiosis, oxidative stress, insulin resistance, and inflammation in MASLD mice. Transcriptomic analysis and molecular data revealed that NP-NAR promoted fatty acid oxidation via activation of the PPAR signaling pathway, reduced hepatic lipid uptake and lipogenesis by inhibiting the expressions of key genes including CD36, ACC, and FASN. Moreover, NP-NAR modulated cholesterol metabolism by inhibiting the classical bile acid synthesis pathway. 16 S rDNA gene sequencing revealed a disbalanced gut microbiota in MASLD mice, whereas NP-NAR treatment statistically reversed the abundance changes of several intestinal bacteria at the phylum and genus levels, which partly contributed to the balance in intestinal metabolite production, including short-chain fatty acids. In conclusion, these findings suggest that NP-NAR may be a promising candidate for the treatment of obesity-associated MASLD, offering new insight into the mechanisms underlying NAR's efficacy against MASLD.
Collapse
Affiliation(s)
- Lu Dong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China
| | - Wenyong Lou
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China
| | - Congfei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Juan Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong Province, 510641, China.
| |
Collapse
|