1
|
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
|
2
|
Yang X, Yan H, Guo R, Chen Y. Association between the NHHR and hepatic steatosis and liver fibrosis: a population-based study. Sci Rep 2025; 15:8462. [PMID: 40069258 PMCID: PMC11897350 DOI: 10.1038/s41598-025-90818-2] [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: 12/18/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is strongly associated with dyslipidemia, and the non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) is a more comprehensive indicator of lipids. This study aimed to investigate the association between NHHR and hepatic steatosis and liver fibrosis. The 2017-2020 national health and nutrition examination survey (NHANES) dataset was used for the cross-sectional survey. NHHR was calculated by lipid profiling, and the controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) were determined by vibration-controlled transient elastography (VCTE). Multiple linear regression models were used to test the linear association between NHHR and hepatic steatosis and liver fibrosis. Fitted smoothing curves and threshold effect analysis were used to describe the nonlinear relationships. This population-based study included 6575 adults (≥ 18 years). After adjusting for covariates, we found a U-shaped association between NHHR and hepatic steatosis, with a breakpoint of 1.26. There was a negative association on the left side of the breakpoint (OR [95% CI] - 24.31 [- 43.92, - 4.70]) and a positive association on the right side of the breakpoint (OR [95% CI] 3.82 [2.05, 5.59]). There was no significant association between NHHR and liver fibrosis. In addition, subgroup analyses and interaction tests showed stable results. In summary, NHHR has a U-shaped association with hepatic steatosis and no significant association with liver fibrosis. Keeping NHHR below 1.26 may be an effective option to reduce the risk of hepatic steatosis. NHHR is a more efficient and cost-effective marker for NAFLD surveillance that can be utilized in future clinical practice.
Collapse
Affiliation(s)
- Xiaoxian Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haiyi Yan
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Playground, Haidian District, Beijing, China
| | - Rui Guo
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Playground, Haidian District, Beijing, China
| | - Yan Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Playground, Haidian District, Beijing, China.
| |
Collapse
|
3
|
Xiong FX, Sun L, Zhang XJ, Chen JL, Zhou Y, Ji XM, Meng PP, Wu T, Wang XB, Hou YX. Machine learning-based models for advanced fibrosis in non-alcoholic steatohepatitis patients: A cohort study. World J Gastroenterol 2025; 31:101383. [DOI: 10.3748/wjg.v31.i9.101383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/02/2024] [Accepted: 01/08/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND The global prevalence of non-alcoholic steatohepatitis (NASH) and its associated risk of adverse outcomes, particularly in patients with advanced liver fibrosis, underscores the importance of early and accurate diagnosis.
AIM To develop a machine learning-based diagnostic model for advanced liver fibrosis in NASH patients.
METHODS A total of 749 patients who underwent liver biopsy at Beijing Ditan Hospital, Capital Medical University, between January 2010 and January 2020 were included. Patients were randomly divided into training (n = 522) and validation (n = 224) cohorts. Five machine learning models were applied to predict advanced liver fibrosis, with feature selection based on Shapley Additive Explanations (SHAP). The diagnostic performance of these models was compared to traditional scores such as the aspartate aminotransferase to platelet ratio index (APRI) and fibrosis index based on the 4 factors (FIB-4), using metrics including the area under the receiver operating characteristic curve (AUROC), decision curve analysis (DCA), and calibration curves.
RESULTS The Extreme Gradient Boosting (XGBoost) model outperformed all other machine learning models, achieving an AUROC of 0.934 (95%CI: 0.914-0.955) in the training cohort and 0.917 (95%CI: 0.880-0.953) in the validation cohort (P < 0.001). Incorporating liver stiffness measurement into the model further improved its performance, with an AUROC of 0.977 (95%CI: 0.966-0.980) in the training cohort and 0.970 (95%CI: 0.950-0.990) in the validation cohort, significantly surpassing APRI and FIB-4 scores (P < 0.001). The XGBoost model also demonstrated superior clinical utility, as evidenced by DCA and calibration curve analysis in both cohorts.
CONCLUSION The XGBoost model provides a highly accurate, non-invasive diagnosis of advanced liver fibrosis in NASH patients, outperforming traditional methods. An online tool based on this model has been developed to assist clinicians in evaluating the risk of advanced liver fibrosis.
Collapse
Affiliation(s)
- Fei-Xiang Xiong
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Lei Sun
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Department of Pathology, Beijing Ditan Hospital, Beijing 100015, China
| | - Xue-Jie Zhang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jia-Liang Chen
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yang Zhou
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiao-Min Ji
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Pei-Pei Meng
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Tong Wu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xian-Bo Wang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yi-Xin Hou
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| |
Collapse
|
4
|
Karimzadeh K, Unniappan S, Zahmatkesh A. Spirulina platensis Peptide-Loaded Nanoliposomes Alleviate Hepatic Lipid Accumulation in Male Wistar Rats by Influencing Redox Homeostasis and Lipid Metabolism via the AMPK Signaling Pathway. Appl Biochem Biotechnol 2025; 197:1696-1725. [PMID: 39601973 DOI: 10.1007/s12010-024-05089-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] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Spirulina platensis low-molecular-weight peptides (SP) have been reported to exhibit antioxidant and hepatoprotective properties. However, the limited bioavailability and solubility of SPs limit their potential applications. In this study, to examine the potential anti-obesity effects and underlying mechanisms of SPs, high-fat diet-induced non-alcoholic fatty liver disease (NAFLD) model rats were treated with SPs and SP-loaded nanoliposomes. Furthermore, hepatic biochemical parameters, inflammatory markers, histopathological changes, and genes involved in AMPK signaling were analyzed. SP-loaded nanoliposomes demonstrated a spherical shape with slower and sustained SP release. SP and SP-loaded nanoliposomes mitigated hepatic damage by lowering serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and increasing hepatic antioxidant enzymes, which are manifested in improving histopathological findings. In addition, notably, SP-loaded nanoliposomes downregulated lipogenic fatty acid synthase (FAS) and sterol regulatory element-binding protein-1c (SREBP-1c) in the liver. Meanwhile, an upregulation of phosphorylated AMP-activated protein kinase (P-AMPK), lipid acid oxidation-related genes carnitine palmitoyltransferase-1 (CPT-1), and peroxisome proliferator-activated receptor alpha (PPAR-α) was found in the rat liver. This data implies that SP and SP-loaded nanoliposomes exhibit protective potential in rats against the HFD-induced NAFLD, which is mediated through the activation of the AMPK signaling pathway.
Collapse
Affiliation(s)
- Katayoon Karimzadeh
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada.
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Asgar Zahmatkesh
- Aquaculture Department, Gilan Agricultural and Natural Resources Research and Education Center, AREEO, Rasht, Iran
| |
Collapse
|
5
|
Zúñiga-Hernández J, Farias C, Espinosa A, Mercado L, Dagnino-Subiabre A, Campo AD, Illesca P, Videla LA, Valenzuela R. Modulation of Δ5- and Δ6-desaturases in the brain-liver axis. Nutrition 2025; 131:112629. [PMID: 39642695 DOI: 10.1016/j.nut.2024.112629] [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: 05/04/2024] [Revised: 10/04/2024] [Accepted: 10/30/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Obesity is associated with liver depletion of ω-3 polyunsaturated fatty acids (ω-3 PUFAS) promoting steatosis and inflammation, whose levels are maintained by diet or biosynthesis involving Δ-5D, Δ-6D desaturases and elongases. METHOD We aimed to assess Δ-5D and Δ-6D activities in liver and brain from mice fed a control diet (CD) or high-fat diet (HFD) for four to sixteen weeks. RESULTS HFD led to (1) an early (4 weeks) enhancement in liver Δ-5D, Δ-6D, and PPAR-α activities, without changes in oxidative stress, liver damage or fat accumulation; (2) a latter progressive loss in hepatic desaturation with insufficient compensatory increases in mRNA and protein expression, leading to ω-3 PUFA depletion, PPAR-α down-regulation reducing FA oxidation, and liver steatosis with enhancement in lipogenesis; and (3) brain ω-3 PUFA depletion after 12 to 16 weeks of HFD feeding. CONCLUSION In conclusion, the brain-liver axis is drastically affected by obesity in a time dependent fashion.
Collapse
Affiliation(s)
| | - Camila Farias
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Espinosa
- Escuela de Medicina, Campus San Felipe, Universidad de Valparaíso, San Felipe, Chile; Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Lorena Mercado
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile; Direccion de postgrado, Facultad Medicina, Universidad Andres Bello, Santiago, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, CIESAL, Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Escuela de Química y Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paola Illesca
- Laboratorio de Estudio de Enfermedades Metabólicas Relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
6
|
Bayram S, Ülger Y. Association of PNPLA3 rs738409 C > G and rs2896019 T > G Polymorphisms with Nonalcoholic Fatty Liver Disease in a Turkish Population from Adıyaman Province. Genet Test Mol Biomarkers 2025; 29:63-73. [PMID: 40101239 DOI: 10.1089/gtmb.2024.0481] [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: 03/20/2025] Open
Abstract
Objectives: The purpose of this study was to investigate the effect of patatin-like phospholipase domain-containing protein 3 (PNPLA3) rs738409 C > G and rs2896019 T > G polymorphisms on genetic susceptibility to nonalcoholic fatty liver disease (NAFLD) in a Turkish population from Adıyaman province, located in the Southeast Anatolia Region of Turkey. Materials and Methods: This hospital-based molecular epidemiological case-control study analyzed the PNPLA3 rs738409 C > G and rs2896019 T > G polymorphisms in 335 NAFLD cases and 410 healthy controls. Genotype frequencies were determined using real-time polymerase chain reaction with the TaqMan assay. The association with NAFLD susceptibility was evaluated by calculating odds ratios (ORs) and 95% confidence intervals (CIs) using an unconditional logistic regression model. Results: We found that the PNPLA3 rs738409 C > G (CC vs. GG: OR = 1.90, 95% CI = 1.05-3.44) and rs2896019 T > G (TT vs. GG: OR = 3.24, 95% CI = 1.44-7.27) polymorphisms were linked to an increased risk of NAFLD in almost all genetic models (p < 0.05). In addition, the PNPLA3 Grs738409/Grs2896019 haplotype was associated with NAFLD development (p < 0.05). Significant differences in alanine aminotransferase and aspartate aminotransferase enzyme levels were observed across the genotypes of these polymorphisms (p < 0.05). Conclusion: This is the first study on PNPLA3 single nucleotide polymorphisms (SNPs) and NAFLD in the Turkish population of Adıyaman Province, Southeast Anatolia. Our findings suggest that the PNPLA3 rs738409 C > G and rs2896019 T > G polymorphisms, along with their haplotypes, may influence NAFLD susceptibility. Further independent studies with larger sample sizes and diverse populations are needed to confirm these results.
Collapse
Affiliation(s)
- Süleyman Bayram
- Faculty of Health Sciences, Department of Public Health Nursing, Adıyaman University, Adıyaman, Turkey
| | - Yakup Ülger
- Faculty of Medicine, Department of Gastroenterology, Çukurova University, Adana, Turkey
| |
Collapse
|
7
|
Ilha M, Sehgal R, Matilainen J, Rilla K, Kaminska D, Gandhi S, Männistö V, Ling C, Romeo S, Pajukanta P, Pirinen E, Virtanen KA, Pietiläinen KH, Vaittinen M, Pihlajamäki J. Indole-3-propionic acid promotes hepatic stellate cells inactivation. J Transl Med 2025; 23:253. [PMID: 40025530 PMCID: PMC11871697 DOI: 10.1186/s12967-025-06266-z] [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: 10/09/2024] [Accepted: 02/16/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND & AIMS We have previously reported that the serum levels of gut-derived tryptophan metabolite indole-3-propionic acid (IPA) are lower in individuals with liver fibrosis. Now, we explored the transcriptome and DNA methylome associated with serum IPA levels in human liver from obese individuals together with IPA effects on shifting the hepatic stellate cell (HSC) phenotype to inactivation in vitro. METHODS A total of 116 obese individuals without type 2 diabetes (T2D) (age 46.8 ± 9.3 years; BMI: 42.7 ± 5.0 kg/m2) from the Kuopio OBesity Surgery (KOBS) study undergoing bariatric surgery were included. Circulating IPA levels were measured using LC-MS, liver transcriptomics with total RNA-sequencing and DNA methylation with Infinium HumanMethylation450 BeadChip. Human hepatic stellate cells (LX-2) where used for in vitro experiments. RESULTS Serum IPA levels were associated with the expression of liver genes enriched for apoptosis, mitophagy and longevity pathways in the liver. AKT serine/threonine kinase 1 (AKT1) was the shared and topmost interactive gene from the liver transcript and DNA methylation profile. IPA treatment induced apoptosis, reduced mitochondrial respiration as well as modified cell morphology, and mitochondrial dynamics by modulating the expression of genes known to regulate fibrosis, apoptosis, and survival in LX-2 cells. CONCLUSION In conclusion, these data support that IPA has a plausible therapeutic effect and may induce apoptosis and the HSC phenotype towards the inactivation state, extending the possibilities to suppress hepatic fibrogenesis by interfering with HSC activation and mitochondrial metabolism.
Collapse
Affiliation(s)
- Mariana Ilha
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, USA
| | - Ratika Sehgal
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany
| | - Johanna Matilainen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Division of Cardiology, UCLA, Los Angeles, CA, USA
| | - Shrey Gandhi
- Institute of Immunology, University of Münster, Münster, Germany
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster, Germany
| | - Ville Männistö
- Departments of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, Göteborg, Sweden
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Institute for Precision Health, School of Medicine, UCLA, Los Angeles, CA, USA
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Research Unit for Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Obesity Center, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Maija Vaittinen
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
8
|
Ye M, He Y, Xia Y, Zhong Z, Kong X, Zhou Y, Xia W, Wang W, Fan H, Chen L, Wu X, Li Q. Association Between Serum Zinc and Non-Alcoholic Fatty Liver Disease and Advanced Liver Fibrosis: NHANES 2011-2016. Biol Trace Elem Res 2025; 203:1305-1316. [PMID: 38861177 DOI: 10.1007/s12011-024-04261-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
Limited and inconclusive evidence exists regarding the correlation between serum zinc levels and non-alcoholic fatty liver disease (NAFLD) and advanced fibrosis. The objective of this cross-sectional study was to investigate the association between serum zinc concentration and both NAFLD and advanced liver fibrosis among the United States (US) adults. 3398 subjects from National Health and Nutrition Examination Survey (NHANES) 2011-2016 were included. Serum zinc concentration was measured by inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS). NAFLD was diagnosed with Hepatic Steatosis Index (HSI), and advanced fibrosis risk was assessed by NAFLD Fibrosis Score (NFS). Weighted logistic regression and restricted cubic splines (RCS) were used to examine the association between serum zinc concentration and NAFLD and advanced fibrosis. Linear trend tests were conducted by incorporating the median of serum zinc quartiles as a continuous variable in the models. We employed sensitivity analysis and subgroup analysis to enhance the robustness of our results. The results from the RCS regression revealed no evident nonlinear relationship between serum zinc concentration and the presence of NAFLD and advanced fibrosis (p-nonlinear > 0.05). Compared with those in the lowest quartile (Q1) of serum zinc concentrations, the odds ratios (95% confidence intervals) of NAFLD were 1.49 (0.89,2.49) in Q2, 0.99 (0.68,1.45) in Q3, and 2.00 (1.40,2.86) in Q4 (p-trend = 0.002). Similarly, the odds ratios (95% confidence intervals) for advanced fibrosis in Q2-4 compared to Q1 were 0.86 (0.50,1.47), 0.60 (0.26,1.39), and 0.41 (0.21,0.77), respectively (p-trend = 0.006). Subgroup analyses and sensitivity analyses reinforce the same conclusion. The investigation revealed a positive linear relationship between serum zinc concentrations and the probability of developing NAFLD. Conversely, an inverse correlation was observed between serum zinc concentrations and the incidence of advanced liver fibrosis among individuals diagnosed with NAFLD.
Collapse
Affiliation(s)
- Miaomin Ye
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yijia He
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yin Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Ziyi Zhong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaocen Kong
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yunting Zhou
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wenqing Xia
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Weiping Wang
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Huan Fan
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Lu Chen
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaohui Wu
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qian Li
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
9
|
Giannakodimos A, Oikonomou E, Pantelidis P, Theofilis P, Katsiki N, Goliopoulou A, Zakynthinos GE, Korakas E, Kalogera V, Banach M, Lampadiari V, Kassi E, Ikonomidis I, Siasos G. Arterial stiffness as a complication of metabolic dysfunction-associated steatotic liver disease: a systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol 2025:1-14. [PMID: 39988816 DOI: 10.1080/17474124.2025.2471871] [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: 12/22/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION The purpose of this systematic review and meta-analysis is to investigate the association of metabolic dysfunction-associated steatotic liver disease (MASLD) with arterial stiffness and enlighten on potential cardiometabolic co-factors. METHODS A literature search in PubMed/Medline, Embase, Scopus, and Web of Science databases was conducted. All the observational studies comparing arterial stiffness indices between adults with Non-alcoholic Fatty Liver Disease (NAFLD), Metabolic Dysfunction Associated-Fatty Liver Disease (MAFLD), or MASLD and apparently healthy individuals with normal liver function were included. Pulse wave velocity (PWV) and augmentation index (AIx) were mainly used as arterial stiffness indices. RESULTS Fourty one unique studies were included in the systematic review, with 27 deemed eligible for meta-analysis. Patients with MASLD had increased carotid-femoral PWV (14 studies, Mean difference (MD): 0.96 m/s, 95% confidence interval (CI) 0.65-1.27, p < 0.001) compared with healthy individuals. This finding was independent from body mass index, triglycerides, high-density lipoprotein, systolic blood pressure, and fasting plasma glucose. Moreover, patients with MASLD had higher brachial-ankle PWV (13 studies, MD: 78.14 cm/s, 95% CI 60.37-95.90, p < 0.001) and AIx (7 studies, MD: 3.85%, 95% CI 0.87-6.82, p = 0.0195) compared with controls. CONCLUSIONS MASLD is correlated with increased arterial stiffness. This relation is unaffected by common cardiometabolic risk factors. REGISTRATION PROSPERO (ID: CRD42023468258).
Collapse
Affiliation(s)
- Alexios Giannakodimos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Athina Goliopoulou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios E Zakynthinos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil Korakas
- 2nd Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Kalogera
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vaia Lampadiari
- 2nd Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ignatios Ikonomidis
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Eslam M, Fan JG, Yu ML, Wong VWS, Cua IH, Liu CJ, Tanwandee T, Gani R, Seto WK, Alam S, Young DY, Hamid S, Zheng MH, Kawaguchi T, Chan WK, Payawal D, Tan SS, Goh GBB, Strasser SI, Viet HD, Kao JH, Kim W, Kim SU, Keating SE, Yilmaz Y, Kamani L, Wang CC, Fouad Y, Abbas Z, Treeprasertsuk S, Thanapirom K, Al Mahtab M, Lkhagvaa U, Baatarkhuu O, Choudhury AK, Stedman CAM, Chowdhury A, Dokmeci AK, Wang FS, Lin HC, Huang JF, Howell J, Jia J, Alboraie M, Roberts SK, Yoneda M, Ghazinian H, Mirijanyan A, Nan Y, Lesmana CRA, Adams LA, Shiha G, Kumar M, Örmeci N, Wei L, Lau G, Omata M, Sarin SK, George J. The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic dysfunction-associated fatty liver disease. Hepatol Int 2025:10.1007/s12072-024-10774-3. [PMID: 40016576 DOI: 10.1007/s12072-024-10774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/28/2024] [Indexed: 03/01/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) affects over one-fourth of the global adult population and is the leading cause of liver disease worldwide. To address this, the Asian Pacific Association for the Study of the Liver (APASL) has created clinical practice guidelines focused on MAFLD. The guidelines cover various aspects of the disease, such as its epidemiology, diagnosis, screening, assessment, and treatment. The guidelines aim to advance clinical practice, knowledge, and research on MAFLD, particularly in special groups. The guidelines are designed to advance clinical practice, to provide evidence-based recommendations to assist healthcare stakeholders in decision-making and to improve patient care and disease awareness. The guidelines take into account the burden of clinical management for the healthcare sector.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of MedicineSchool of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, Kaohsiung Medical University, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China
| | - Ian Homer Cua
- Institute of Digestive and Liver Diseases, St. Luke's Medical Center, Global City, Philippines
| | - Chun-Jen Liu
- Division of Gastroenterology and Hepatology, Department of Internal MedicineHepatitis Research CenterGraduate Institute of Clinical Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tawesak Tanwandee
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rino Gani
- Department of Internal Medicine, Hepatobiliary Division, Dr. Cipto Mangunkusumo National General Hospital, Universitas Indonesia, Pangeran Diponegoro Road No. 71St, Central Jakarta, 10430, Indonesia
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shahinul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Dan Yock Young
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Saeed Hamid
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Payawal
- Department of Medicine, Cardinal Santos Medical Center, Mandaluyong, Philippines
| | - Soek-Siam Tan
- Department of Hepatology, Selayang Hospital, Batu Caves, Malaysia
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Hang Dao Viet
- Internal Medicine Faculty, Hanoi Medical University, Hanoi, Vietnam
| | - Jia-Horng Kao
- Graduate Institute of Clinical MedicineDepartment of Internal MedicineHepatitis Research CenterDepartment of Medical Research, National Taiwan University College of Medicine, National Taiwan University, National Taiwan University Hospital, 1 Chang-Te Street, 10002, Taipei, Taiwan
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | - Chia-Chi Wang
- Buddhist Tzu Chi Medical Foundation and School of Medicine, Taipei Tzu Chi Hospital, Tzu Chi University, Taipei, Taiwan
| | - Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Cairo, Egypt
| | - Zaigham Abbas
- Department of Hepatogastroenterology, Dr.Ziauddin University Hospital, Clifton, Karachi, Pakistan
| | | | | | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Undram Lkhagvaa
- Department of Health Policy, School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Oidov Baatarkhuu
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Ashok Kumar Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, 110070, India
| | | | - Abhijit Chowdhury
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - A Kadir Dokmeci
- Department of Medicine, Ankara University School of Medicine, Ankara, Turkey
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Institute of Clinical Medicine, School of Medicine, Taipei Veterans General Hospital, National Yang-Ming Chiao Tung University, No. 201, Section 2, Shipai RdNo. 155, Section 2, Linong St, Beitou District, Taipei City, 112, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jess Howell
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Clayton, VIC, 3008, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, 3165, Australia
| | - Jidong Jia
- Liver Research Center, Beijing Key Laboratory of Translational Medicine On Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, 11884, Egypt
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Central Clinical School, The Alfred, Monash University, Melbourne, Australia
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hasmik Ghazinian
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Aram Mirijanyan
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Leon A Adams
- Medical School, Faculty of Medicine and Health Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Gamal Shiha
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansoura, Egypt
| | - Manoj Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Necati Örmeci
- Department of Gastroenterohepatology, Istanbul Health and Technology University, Istanbul, Turkey
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - George Lau
- Humanity and Health Medical Group, Humanity and Health Clinical Trial Center, Hong Kong SAR, China
- The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
- University of Tokyo, Tokyo, Japan
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
11
|
Kim HJ, Kim JY, Lee YM, Hong YH, Kang B, Choe BH, Yi DY, Lee EH, Kim SC, Choi YJ, Jang HJ, Choi SY. Association of antinuclear antibody positivity with liver disease severity in pediatric metabolic dysfunction-associated steatotic liver disease. Front Pediatr 2025; 13:1527605. [PMID: 40115315 PMCID: PMC11925201 DOI: 10.3389/fped.2025.1527605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/05/2025] [Indexed: 03/23/2025] Open
Abstract
Background Although antinuclear antibody (ANA) is frequently observed in patients with metabolic dysfunction-associated steatotic liver disease (MASLD), its clinical significance in children remains unclear and controversial. In this study, we investigated the prevalence of ANA positivity and the factors associated with it in pediatric MASLD patients without concurrent autoimmune hepatitis. Methods We retrospectively reviewed the medical records of patients aged 4-18 years diagnosed with MASLD and tested for ANA from January 2015 to December 2020 at 10 hospitals in Korea. All statistical analyses were carried out using SPSS 26.0 and P-values <0.05 were considered statistically significant. Results Out of the 439 patients included, ANAs were present in 89 (20.3%); 51 (57.3%) patients had ANA titer <1:80; 22 (24.7%), <1:160; 10 (11.2%), <1:320; and 6 (6.7%), <1:640. Compared to ANA-negative patients, aspartate aminotransferase (AST, P = 0.003) and alanine aminotransferase (ALT, P = 0.007) levels were significantly higher in ANA-positive patients. The ALT to Platelet Ratio Index (APRI) score was also associated with the ANA-positive patients (P = 0.005). To predict ANA positivity using APRI, the area under receiver operating characteristic (AUROC) curve was 0.597 (p = 0.004), and the APRI cutoff value of >0.893 could predict ANA, with sensitivity and specificity of 42.7% and 72.9%, respectively. Conclusions ANA positivity in pediatric MASLD is associated with greater liver enzyme elevation and increased risk of fibrosis, highlighting the need for careful monitoring in ANA-positive patients.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Department of Pediatrics, Chungnam National University Hospital, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ju Young Kim
- Department of Pediatrics, Daejeon Eulji Medical Center, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Yoo Min Lee
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Yong Hee Hong
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Ben Kang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Byung-Ho Choe
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dae Yong Yi
- Department of Pediatrics, Chung-Ang University Hospital, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Eun Hye Lee
- Department of Pediatrics, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Republic of Korea
| | - Soon Chul Kim
- Department of Pediatrics, Jeonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - You Jin Choi
- Department of Pediatrics, Inje University, Ilsan Paik Hospital, Inje University College of Medicine, Ilsan, Republic of Korea
| | - Hyo-Jeong Jang
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - So Yoon Choi
- Department of Pediatrics, Kosin Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
| |
Collapse
|
12
|
Pecoraro V, Nascimbeni F, Cuccorese M, Gabrielli F, Fasano T, Trenti T. Diagnostic Accuracy of Golgi Protein 73 (GP73) for Liver Fibrosis Staging in Metabolic Dysfunction-Associated Steatotic Liver Disease: A Scoping Review and Cohort Study. Diagnostics (Basel) 2025; 15:544. [PMID: 40075792 PMCID: PMC11898419 DOI: 10.3390/diagnostics15050544] [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: 12/31/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Golgi protein 73 (GP73) is a transmembrane protein expressed by epithelial cells of the bile duct in the normal liver. High serum levels of GP73 have been detected in patients with acute or chronic liver diseases, MASLD, and its measurement has been suggested as a potential biomarker for liver fibrosis staging. We evaluated the utility of GP73 in the diagnosis of MASLD, MASH, and for liver fibrosis staging. Methods: We performed a literature scoping review to map the current evidence about the accuracy of GP73 in patients with MASLD. We searched in Medline and EMBASE for English studies reporting an AUC value of GP73 in diagnosing MASLD and MASH and evaluating GP73 for fibrosis staging. A narrative synthesis of the evidence was conducted. Moreover, we performed an observational study including 84 patients with MASLD, of which 60 were biopsy-confirmed MASH, and different liver fibrosis stages, and 15 healthy controls. Serum GP73 levels were determined using a chemiluminescent assay and reported as mean and standard deviation (SD). Sensitivity (SE), specificity (SP), the area under the receiver operating characteristic (AUROC) curve, and the optimal cut-off value were calculated. Data were considered statistically significant when p < 0.05. Results: Available studies evaluating GP73 in MASLD reported the ability to discriminate MASH from simple steatosis and distinguish patients at different fibrotic stages, but the evidence is still scarce. Our experimental study showed that the serum levels of GP73 were 30 ± 12 ng/mL in MASLD and 32 ± 12 ng/mL in MASH patients and were statistically higher than those of the control group (19 ± 30 ng/mL), increasing from liver fibrosis stage F0 to F4. GP73 levels were significantly higher in patients with significant and advanced fibrosis than controls and no significant fibrosis (p > 0.05). ROC analysis demonstrated that serum GP73 had a good diagnostic potential for MASLD (AUROC 0.85; SE 90%; SP 73%), MASH (AUROC 0.75; SE 82%; SP64%), and significant fibrosis (AUROC 0.7; SE 56%; SP 79%) and was better than other biomarkers for chronic liver diseases. Conclusions: Serum GP73 could support clinicians in the evaluation of patients with MASH and significant fibrosis.
Collapse
Affiliation(s)
- Valentina Pecoraro
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | | - Michela Cuccorese
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | | - Tommaso Fasano
- Complex Structure of Laboratory Medicine, Department of Laboratory Medicine and Pathological Anatomy, AUSL Modena, 41121 Modena, Italy
| | | |
Collapse
|
13
|
Choque Vargas C, Cáceres F, Landeira G, Perez S, Marchi L, Ruffillo G, Tevez S, Puga-Tejada M, Fassio E. Cardiovascular events and incident diabetes in 220 patients with MASLD according to basal liver fibrosis: a 10-year follow-up historic cohort. Eur J Gastroenterol Hepatol 2025:00042737-990000000-00490. [PMID: 39975992 DOI: 10.1097/meg.0000000000002943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
AIM The aim of this study is to analyze association between liver fibrosis with CVE, incident diabetes, and cirrhosis complications. METHODS Historic cohort of biopsy-proven MASLD patients, divided into two groups: F0-F2 vs F3-F4 fibrosis. Baseline data included metabolic traits and liver function tests. Patients were contacted and scheduled for laboratory analysis and elastography. Endpoints were (a) CVE, defined as any of acute myocardial infarction, coronary stenting, ischemic cardiopathy, and stroke; (b) incident diabetes; (c) cirrhosis complications. Baseline data were collected at the time of liver biopsy, while follow-up data were recovered through personal interview or medical records. A stepwise logistic regression determined predictive variables for each endpoint. RESULTS Study population included 220 patients with median age 53 years, and 145 were women; baseline fibrosis was F0-F2 in 165 patients and F3-F4 in 55 patients; median follow-up was 9.9 years. A higher percentage of F3-F4 patients had CVE (29.4%) than F0-F2 ones (13.1%) (hazard ratio 2.42; 95% CI: 1.26-4.6; P = 0.008). Incident diabetes occurred in 53.3% of F3-F4 and 20.2% of F0-F2 cohort (hazard ratio 3.04; 95% CI: 1.99-4.86; P < 0.001); cirrhosis complications occurred in 9/55 F3-F4 patients and in 1/165 F0-F2 ones (hazard ratio 26.3; 95% CI: 3.3-208.3; P = 0.002). Multivariate analysis confirmed liver fibrosis as an independent predictor of incident diabetes and cirrhosis complications. CVE were associated with baseline diabetes and aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratio. CONCLUSION In a cohort of 220 MASLD patients followed for 9.9 years, baseline F3-F4 was associated with incident diabetes and cirrhosis complications. AST/ALT ratio and diabetes were associated with CVE.
Collapse
Affiliation(s)
- Cinthia Choque Vargas
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Francisco Cáceres
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Graciela Landeira
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Soledad Perez
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Laura Marchi
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Gabriela Ruffillo
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Silvina Tevez
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| | - Miguel Puga-Tejada
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
- División de Investigación Médica & Bioestadística, Instituto Ecuatoriano de Enfermedades Digestivas, Guayaquil, Ecuador
| | - Eduardo Fassio
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Prof. Alejandro Posadas, El Palomar, Buenos Aires, Argentina
| |
Collapse
|
14
|
Stegmann SK, Vohlen C, Im NG, Niehues J, Selle J, Janoschek R, Kuiper-Makris C, Lang S, Demir M, Steffen HM, Quaas A, Lackmann JW, Nierhoff D, Neumann-Haefelin C, Dötsch J, Alejandre Alcazar MA, Kasper P. Perinatal obesity primes the hepatic metabolic stress response in the offspring across life span. Sci Rep 2025; 15:6416. [PMID: 39984579 PMCID: PMC11845730 DOI: 10.1038/s41598-025-90082-4] [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: 09/26/2024] [Revised: 12/22/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
Perinatal obesity is associated with an increased risk of metabolic diseases and hepatic dysfunction in offspring. However, the underlying mechanisms of this metabolic programming remain incompletely understood. This study aimed to elucidate the influence of maternal obesity and early life exposure to high-fat diet on offspring liver phenotype, hepatokine profile, and key components of hepatic metabolism. To this end, we employed a murine high-fat diet-induced perinatal obesity model, investigating the offspring in early life and late adulthood. After exposure to perinatal obesity, the offspring showed a significantly increased body weight in early life with no histological signs of steatosis, but a dysregulated hepatokine profile. Proteomic profiling, followed by molecular analyses, revealed a decreased lipogenesis and increased fatty acid oxidation, suggesting a protective mechanism against the development of steatosis. These changes were accompanied by increased markers of lipid peroxidation and DNA damage, indicating increased oxidative stress. Concomitantly, the antioxidative enzymes catalase and superoxide dismutase 2 were significantly reduced and oxidative phosphorylation was impaired, implying an altered oxidative stress response. While changes in oxidative stress level were only detected in early life, the lipid metabolism was altered across life span. This metabolic programming could determine the resilience and susceptibility to chronic liver disease later in life.
Collapse
Affiliation(s)
- Sarah K Stegmann
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Nam Gyu Im
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jana Niehues
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jaco Selle
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Celien Kuiper-Makris
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sonja Lang
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité University Medicine Berlin, Berlin, Germany
| | - Hans-Michael Steffen
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Department of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan-Wilm Lackmann
- Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Dirk Nierhoff
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Christoph Neumann-Haefelin
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Kasper
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
15
|
Athira AS, Abhijith B, Sruthi PK, Ragavamenon AC, Lankalapalli RS, Reshma MV. Lyophilized ash gourd ( Benincasa hispida (Thunb.) Cogn.) juice alleviates diet-induced prediabetes in a rat model. Food Funct 2025; 16:1534-1549. [PMID: 39903217 DOI: 10.1039/d4fo05327c] [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: 02/06/2025]
Abstract
Prediabetes is characterized by elevated blood sugar levels, indicating an increased risk of developing diabetes. This study evaluated the effects of ash gourd (AG), a tropical fruit from the Cucurbitaceae family, on prediabetes, as well as its phytochemical composition. A prediabetic rat model was developed in Sprague-Dawley (SD) rats by administering a high fat diet (HFD) for 16 weeks. This model exhibited reduced pancreatic function, heightened insulin resistance, and decreased insulin sensitivity compared to a standard diet group, leading to hyperglycemia and dyslipidemia, hallmarks of prediabetic conditions. Histological analysis of hepatic tissue revealed macro- and microvesicular fat accumulation and inflammatory changes, supporting these findings. This study highlights the utility of HFD-induced SD rats as a model for prediabetic conditions. Following this, lyophilized ash gourd juice (LAGJ) powder was administered to the prediabetic rat model to assess its potential for reversing prediabetic conditions. LAGJ administration resulted in a significant reduction in fasting blood sugar (FBS) levels, glucose intolerance, and insulin resistance. Additionally, LAGJ significantly mitigated fatty liver changes compared to the prediabetic untreated control (PUC) group. Histological examination of liver tissue in the LAGJ treated group showed a typical architecture similar to that of the normal control group. These findings indicate that LAGJ could be a promising intervention for individuals with prediabetes who are at risk of developing type 2 diabetes and fatty liver disease. Phytochemical analysis of AG pulp revealed the presence of stigmasterol, stigmasteryl β-glucoside, and 6'-O-palmitoyl stigmasteryl β-glucoside.
Collapse
Affiliation(s)
- A S Athira
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Balan Abhijith
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - P K Sruthi
- Department of Biochemistry, Amala Cancer Research Centre, Amala Nagar, Thrissur-680555, Kerala, India
| | - Achuthan C Ragavamenon
- Department of Biochemistry, Amala Cancer Research Centre, Amala Nagar, Thrissur-680555, Kerala, India
| | - Ravi S Lankalapalli
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram 695019, Kerala, India
| | - M V Reshma
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
16
|
Lajeunesse-Trempe F, Dugas S, Maltais-Payette I, Tremblay ÈJ, Piché ME, K. Dimitriadis G, Lafortune A, Marceau S, Biertho L, Tchernof A. Anthropometric Indices and Metabolic Dysfunction-Associated Fatty Liver Disease in Males and Females Living With Severe Obesity. Can J Gastroenterol Hepatol 2025; 2025:5545227. [PMID: 39989658 PMCID: PMC11847611 DOI: 10.1155/cjgh/5545227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/28/2024] [Accepted: 11/07/2024] [Indexed: 02/25/2025] Open
Abstract
Introduction: Metabolic dysfunction-associated fatty liver disease (MAFLD) is highly prevalent among people living with severe obesity (body mass index [BMI] ≥ 35 kg/m2). However, it remains unknown how sex and adipose tissue distribution are related to MAFLD onset and progression into metabolic dysfunction-associated steatohepatitis (MASH) or advanced stages of fibrosis. Methodology: We retrospectively studied patients with severe obesity who were eligible for bariatric surgery. Demographic characteristics, biomarkers, and cardiometabolic comorbidities were reported. Anthropometric indices such as BMI, waist circumference (WC), waist-to-hip ratio (WHR), waist-to-height ratio (WHtR), neck circumference (NC), lipid accumulation product (LAP), visceral adiposity index (VAI), body adiposity index (BAI), abdominal volume index (AVI), and body roundness index (BRI) were measured or calculated. MAFLD, MASH, and stages of fibrosis (F1-F4) were established from perioperative liver biopsies. Standardized univariate and multivariate logistic regression analyses were used to examine the association between demographic variables, anthropometric indices, cardiometabolic conditions, and the risk of MASH or severe fibrosis (F2-F4). Results: A total of 2091 participants with severe obesity were included in the analyses; BMI 47.9 ± 7.3 kg/m2, age 46.2 ± 11.2 years, and 68.4% females. Overall, MAFLD prevalence was 79.5%, with 44.5% having MASH and 24.4% having severe fibrosis (Stage 2 or higher). No anthropometric indices of adiposity were associated with MASH or fibrosis severity. In this population, female sex was a risk factor for severe fibrosis (OR: 1.27, 95% CI 1.01-1.59, p < 0.05). Conclusions: MAFLD and MASH are highly prevalent in individuals living with severe obesity, but no anthropometric indices or laboratory tests are good predictors of MAFLD or MASH in this population. When MAFLD is diagnosed, our results suggest that females with severe obesity might be at higher risk of advanced stages of fibrosis.
Collapse
Affiliation(s)
- Fannie Lajeunesse-Trempe
- Department of Specialized Medicine, Internal Medicine, Quebec Heart and Lung Institute, Laval University, Quebec City, Quebec, Canada
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
- Faculty of Life Sciences and Medicine, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - Selena Dugas
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Ina Maltais-Payette
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Ève-Julie Tremblay
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| | - Marie-Eve Piché
- Department of Medicine, Laval University, Quebec City, Quebec, Canada
| | - Georgios K. Dimitriadis
- Faculty of Life Sciences and Medicine, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
- Department of Endocrinology, King's College Hospital NHS Foundation Trust, London, UK
| | - Annie Lafortune
- Department of Surgery, Laval University, Quebec City, Quebec, Canada
| | - Simon Marceau
- Department of Surgery, Laval University, Quebec City, Quebec, Canada
| | - Laurent Biertho
- Department of Surgery, Laval University, Quebec City, Quebec, Canada
| | - André Tchernof
- Faculty of Agriculture and Food Sciences, School of Nutrition, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
17
|
Lin L, Lai J, Luo L, Ye J, Zhong B. Ethnic differences in metabolic and histologic features among White, Hispanic, Black and Asian patients with metabolic-associated Steatotic liver disease: A network meta-analysis. Ann Hepatol 2025; 30:101780. [PMID: 39952324 DOI: 10.1016/j.aohep.2025.101780] [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: 11/12/2023] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 02/17/2025]
Abstract
INTRODUCTION AND OBJECTIVES Current evidence on the impact of ethnic disparities on metabolic-associated steatotic liver disease (MASLD) is limited to individual studies with small sample sizes from specific regions. This network meta-analysis aimed to assess variations in metabolism and histological characteristics of MASLD among four ethnicities. MATERIALS AND METHODS Observational studies on MASLD involving at least two ethnic groups (White, Black, Asian, and Hispanic) were identified from PubMed, Embase, and Web of Science databases up to May 7th, 2024, for inclusion in this study. The results were reported as unstandardized mean differences (MDs) and odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS A total of twenty-seven articles involving 14,440 non-Hispanic Whites, 4,927 non-Hispanic Blacks, 5,254 Asians, and 8,344 Hispanic MASLD patients were included in this study. The prevalence of type 2 diabetes mellitus of all ethnic groups combined was 33%, without significant difference among the four ethnicities. Asians showed higher levels of total cholesterol compared to the other groups, while Blacks had the lowest levels of alanine aminotransferase. Among biopsy-proven MASLD patients, Blacks individuals had a lower risk of significant fibrosis compared to Whites (OR=0.63, 95% CI: 0.45 to 0.87), as well as lower risks of liver inflammation (OR=0.53, 95% CI: 0.29 to 0.95) and nonalcoholic steatohepatitis (NASH) (OR=0.53, 95% CI: 0.29 to 0.95) compared to Hispanics. CONCLUSIONS Asians MASLD patients had higher risk of suffering from abnormal lipid metabolism while Black MASLD patients presented milder liver histologic features than both Whites and Hispanics individuals.
Collapse
Affiliation(s)
- Limin Lin
- Department of Gastroenterology of the First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China
| | - Jiaming Lai
- Department of Gastroenterology of the First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China
| | - Ling Luo
- Department of Gastroenterology of the First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China
| | - Junzhao Ye
- Department of Gastroenterology of the First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China.
| | - Bihui Zhong
- Department of Gastroenterology of the First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China.
| |
Collapse
|
18
|
Lam WLM, Gabernet G, Poth T, Sator-Schmitt M, Oquendo MB, Kast B, Lohr S, de Ponti A, Weiß L, Schneider M, Helm D, Müller-Decker K, Schirmacher P, Heikenwälder M, Klingmüller U, Schneller D, Geisler F, Nahnsen S, Angel P. RAGE is a key regulator of ductular reaction-mediated fibrosis during cholestasis. EMBO Rep 2025; 26:880-907. [PMID: 39747668 PMCID: PMC11811172 DOI: 10.1038/s44319-024-00356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Ductular reaction (DR) is the hallmark of cholestatic diseases manifested in the proliferation of bile ductules lined by biliary epithelial cells (BECs). It is commonly associated with an increased risk of fibrosis and liver failure. The receptor for advanced glycation end products (RAGE) was identified as a critical mediator of DR during chronic injury. Yet, the direct link between RAGE-mediated DR and fibrosis as well as the mode of interaction between BECs and hepatic stellate cells (HSCs) to drive fibrosis remain elusive. Here, we delineate the specific function of RAGE on BECs during DR and its potential association with fibrosis in the context of cholestasis. Employing a biliary lineage tracing cholestatic liver injury mouse model, combined with whole transcriptome sequencing and in vitro analyses, we reveal a role for BEC-specific Rage activity in fostering a pro-fibrotic milieu. RAGE is predominantly expressed in BECs and contributes to DR. Notch ligand Jagged1 is secreted from activated BECs in a Rage-dependent manner and signals HSCs in trans, eventually enhancing fibrosis during cholestasis.
Collapse
Affiliation(s)
- Wai-Ling Macrina Lam
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Gisela Gabernet
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tanja Poth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Melanie Sator-Schmitt
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Morgana Barroso Oquendo
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Bettina Kast
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Sabrina Lohr
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Aurora de Ponti
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Lena Weiß
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Martin Schneider
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Helm
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Müller-Decker
- Tumor Models Unit, Center for Preclinical Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ursula Klingmüller
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Doris Schneller
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Fabian Geisler
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, München, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
19
|
Wang TJ, Jirapinyo P, Shah R, Schuster K, Papke DJ, Thompson CC, Doyon L, Lautz DB, Ryou M. EUS-guided shear wave elastography for fibrosis screening in patients with obesity and metabolic dysfunction-associated steatotic liver disease: a pilot study (with video). Gastrointest Endosc 2025; 101:456-462.e1. [PMID: 39481576 DOI: 10.1016/j.gie.2024.10.054] [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: 03/10/2024] [Revised: 07/13/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND AND AIMS Liver fibrosis staging is challenging in patients with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). Liver biopsies are invasive, whereas noninvasive tests such as vibration-controlled transient elastography (VCTE) can be inaccurate in patients with obesity. We hypothesized that EUS-guided shear wave elastography (EUS-SWE) is more accurate for liver fibrosis staging in patients with MASLD and obesity; the aim of this pilot study was to test this hypothesis and establish optimal fibrosis stage cutoffs for EUS-SWE. METHODS This was a multicenter, cross-sectional study from prospectively collected data. Consecutive patients who underwent EUS-SWE with subsequent liver biopsy were included. EUS-SWE was compared with Fibrosis-4 Index (FIB-4) and VCTE. Area under the receiver-operating characteristic (AUROC) curve analysis was performed, and 90% sensitivity and specific cutoffs were calculated to determine optimal cutoffs. RESULTS Sixty-two patients were included. Mean body mass index was 40.74 kg/m2. EUS-SWE was superior to FIB-4 in discriminating significant fibrosis (F2; AUROC, .87 vs .61; P < .0048) and advanced fibrosis (F3; AUROC, .93 vs .63; P < .0001), but not cirrhosis (F4; AUROC, .95 vs .81; P = .099). EUS-SWE was superior to VCTE in predicting advanced fibrosis and cirrhosis (P = .0067 and P = .0022, respectively). The 90% sensitivity cutoffs for EUS-SWE were 7.50, 8.48, and 11.30 for F2, F3, and F4, and the 90% specificity cutoffs were 9.82, 10.20, and 14.60. CONCLUSIONS In this pilot study, EUS-SWE was superior to FIB-4 and VCTE for liver fibrosis staging in patients with MASLD and obesity. (Clinical trial registration number: NCT05728697.).
Collapse
Affiliation(s)
- Thomas J Wang
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Pichamol Jirapinyo
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Raj Shah
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts, USA; Division of Gastroenterology, Hepatology, and Nutrition, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Kimberly Schuster
- Center for Weight Loss, Emerson Hospital, Concord, Massachusetts, USA
| | - David J Papke
- Hepatology and Endoscopy, and Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Laura Doyon
- Center for Weight Loss, Emerson Hospital, Concord, Massachusetts, USA
| | - David B Lautz
- Center for Weight Loss, Emerson Hospital, Concord, Massachusetts, USA
| | - Marvin Ryou
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
20
|
Yamaguchi R, Oda T, Nagashima K. Comparison of the diagnostic accuracy of shear wave elastography with transient elastography in adult nonalcoholic fatty liver disease: a systematic review and network meta-analysis of diagnostic test accuracy. Abdom Radiol (NY) 2025; 50:734-746. [PMID: 39240377 PMCID: PMC11794403 DOI: 10.1007/s00261-024-04546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE To compare the diagnostic test accuracy (DTA) of shear wave elastography (SWE) to that of transient elastography (TE) for liver fibrosis grade assessment in nonalcoholic fatty liver disease adults. METHODS MEDLINE, The Cochrane Library, and Web of Science were searched. Inclusion criteria were primary studies examining DTA of TE, point SWE (pSWE), two-dimensional SWE (2D-SWE), or magnetic resonance elastography (MRE) with liver biopsy. Network meta-analysis was conducted using a Bayesian bivariate mixed-effects model. RESULTS For fibrosis grade 2 or higher, 15 studies with 25 observations (16 observations for TE, 1 for MRE, 4 for pSWE and 2D-SWE; 2,066 patients) were included; the pooled sensitivity and specificity were 0.79 (95% credible interval (CrI) 0.70-0.86; 95% prediction interval (PI) 0.36-0.96) and 0.73 (95% CrI 0.62-0.82; 95% PI 0.23-0.96) for TE, 0.68 (95% CrI 0.48-0.83; 95% PI 0.23-0.94) and 0.75 (95% CrI 0.53-0.88; 95% PI 0.24-0.97) for pSWE, 0.85 (95% CrI 0.70-0.93; 95% PI 0.40-0.98) and 0.72 (95% CrI 0.49-0.86; 95% PI 0.20-0.96) for 2D-SWE, respectively. The proportion of studies classified as unclear in QUADAS-2 was high, and the results were heterogeneous. CONCLUSION 2D-SWE could be recommended as TE is for liver fibrosis assessment. The protocol of this systematic review and network meta-analysis has been registered in PROSPERO (CRD42022327249). All included primary papers have already been published and the information and data can be used freely.
Collapse
Affiliation(s)
- Ruri Yamaguchi
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aobaku, Sendai, 980-8575, Japan.
| | - Tetsuro Oda
- Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, 160-8582, Japan
- Department of Bioregulation, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8602, Japan
- Division of Cancer Therapeutics, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| |
Collapse
|
21
|
Maltais‐Payette I, Bourgault J, Gauthier M, Biertho L, Marceau S, Julien F, Mitchell PL, Couture C, Brière F, Corbeil J, Arsenault BJ, Tchernof A. Associations between circulating amino acids and metabolic dysfunction-associated steatotic liver disease in individuals living with severe obesity. Physiol Rep 2025; 13:e70171. [PMID: 39868884 PMCID: PMC11770886 DOI: 10.14814/phy2.70171] [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: 10/17/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) describes liver diseases caused by the accumulation of triglycerides in hepatocytes (steatosis) as well as the resulting inflammation and fibrosis. Previous studies have demonstrated that accumulation of fat in visceral adipose tissue compartments and the liver is associated with alterations in the circulating levels of some amino acids, notably glutamate. This study aimed to investigate the associations between circulating amino acids, particularly glutamate, and MASLD. In addition, we hypothesized that liver steatosis, concomitant with visceral adiposity, could contribute to the association between circulating glutamate and visceral obesity. We studied a sample of 150 patients living with severe obesity who were non-diabetic and selected to represent a wide range of MASLD severity. Liver histological features were determined by a pathologist from a biopsy sample obtained at the time of bariatric surgery. Bulk RNA sequencing measured the hepatic mRNA expression level of selected genes related to the urea cycle and glutamate metabolism. Fasting plasma amino acid levels were measured by liquid chromatography coupled with tandem mass spectrometry. Patients with more advanced steatosis had larger visceral adipocytes, higher levels of circulating tyrosine, glutamate, and alanine as well as lower levels of serine. MASLD severity was significantly associated with the hepatic mRNA expression of glutamate metabolism genes such as GLS1, GLUL (positively), and NAGS (inversely). In individuals living with obesity, MASLD severity is associated with visceral adipocyte hypertrophy, higher circulating glutamate as well as potential alterations of hepatic amino acid and nitrogen metabolism.
Collapse
Affiliation(s)
- Ina Maltais‐Payette
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- School of Nutrition, Faculty of Agriculture and Food ScienceLaval UniversityQuebecQuebecCanada
| | - Jérôme Bourgault
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- Department of Molecular Medicine, Faculty of MedicineLaval UniversityQuebecQuebecCanada
| | | | - Laurent Biertho
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- Department of Surgery, Faculty of MedicineLaval UniversityQuebecQuebecCanada
| | - Simon Marceau
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- Department of Surgery, Faculty of MedicineLaval UniversityQuebecQuebecCanada
| | - François Julien
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- Department of Surgery, Faculty of MedicineLaval UniversityQuebecQuebecCanada
| | | | - Christian Couture
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
| | - Francis Brière
- Department of Molecular Medicine, Faculty of MedicineLaval UniversityQuebecQuebecCanada
- Laval University Medical Center – Laval UniversityQuebecQuebecCanada
| | - Jacques Corbeil
- Department of Molecular Medicine, Faculty of MedicineLaval UniversityQuebecQuebecCanada
- Laval University Medical Center – Laval UniversityQuebecQuebecCanada
| | - Benoit J. Arsenault
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- Department of Molecular Medicine, Faculty of MedicineLaval UniversityQuebecQuebecCanada
| | - André Tchernof
- Quebec Heart and Lung Institute – Laval UniversityQuebecQuebecCanada
- School of Nutrition, Faculty of Agriculture and Food ScienceLaval UniversityQuebecQuebecCanada
| |
Collapse
|
22
|
Mocciaro G, George AL, Allison M, Frontini M, Huang‐Doran I, Reiman F, Gribble F, Griffin JL, Vidal‐Puig A, Azzu V, Kay R, Vacca M. Oxidised Apolipoprotein Peptidome Characterises Metabolic Dysfunction-Associated Steatotic Liver Disease. Liver Int 2025; 45:e16200. [PMID: 39822152 PMCID: PMC11740006 DOI: 10.1111/liv.16200] [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: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) encompasses a spectrum of histological conditions ranging from simple steatosis to fibrosing steatohepatitis, and is a risk factor for cardiovascular diseases (CVD). While oxidised apolipoproteins A and B have been linked to obesity and CVD, the association between other oxidised apolipoproteins and MASLD is yet to be established. To fill this gap, we characterised the circulating serum peptidome of patients with MASLD. METHODS We studied the serum of 87 biopsy-confirmed MASLD patients and 20 age- and sex-matched control (CTRL) subjects. We first employed an untargeted LC-MS/MS peptidomics approach (9 CTRL, 32 MASLD) to identify key hits differentially modulated, and subsequently validated the most relevant findings through targeted peptidomics in an enlarged study population (87 MASLD and 20 CTRL). RESULTS Untargeted serum peptidomics identified several oxidised apolipoprotein peptide fragments, including ApoE and ApoC-III, significantly upregulated in MASLD compared to CTRL. Specifically focusing on the oxidative status of intact ApoC-III, studied through its major glycoforms (ApoC-III0, ApoC-IIIi and ApoC-IIIii), we observed a marked reduction in non-oxidised forms of these circulating peptides alongside substantially increased levels of their oxidised proteoforms in MASLD versus controls (but not within the disease stages). Oxidised ApoE and ApoC-III peptide fragments were also significantly correlated with obesity, insulin resistance, dyslipidaemia and transaminases, suggesting a potential link between circulating apolipoprotein oxidation and systemic/hepatic metabolic dysfunction. CONCLUSION Our data reveals a previously unreported oxidised apolipoprotein profile associated with MASLD. The functional and clinical implications of these findings warrant further mechanistic investigation.
Collapse
Grants
- Wellcome Trust
- MR/S010483/1 Medical Research Council
- This research was supported by NIHR Cambridge Biomedical Research Centre (BRC-1215-20014) funding to V.A., M.A., and M.V.The initial establishment of the cohorts was supported by the Evelyn Trust (funding to M.A. and A.V.P.), Mason Medical Research Trust (funding to V.A.), and the Academy of Medical Sciences (funding to V.A.). V.A. was supported by the University of Cambridge. M.A. is supported by Cambridge University Hospitals NHS Foundation Trust. M.V. is supported by the University of Bari (Horizon Europe Seed cod. id. S06-miRNASH), the Foundation for Liver Research (Intramural Funding), Associazione Italiana Ricerca sul Cancro (IG2022 Grant n. 27521) and Ministry of University and Research on Next Generation EU Funds [COD: P202222FCC, CUP: H53D23009960001, D.D. MUR 1366 (01-09-2023), Title: "System Biology" approaches in HCV Patients with Residual Hepatic Steatosis after Viral Eradication; Cod PE00000003, CUP: H93C22000630001, DD MUR 1550, Title: "ON Foods - Research and innovation network on food and nutrition Sustainability, Safety and Security - Working ON Foods"; Cod: CN00000041, CUP: H93C22000430007, Title PNRR "National Center for Gene Therapy and Drugs based on RNA Technology", M4C2-Investment 1.4; Code: CN00000013, CUP: H93C22000450007, Title PNNR: "National Centre for HPC, Big Data and Quantum Computing").A.V.P. is funded by MRC MDU, MRC Metabolic Diseases Unit (MC_UU_00014/5): Disease Model Core, Biochemistry Assay Lab, Histology Core and British Heart Foundation. M.F. is supported by the British Heart Foundation (FS/18/53/33863) and the British Heart Foundation Cambridge Centre for Research Excellence (RE/18/1/34212). This study was supported by the National Institute for Health and Care Research Exeter Biomedical Research Centre. We would like to thank all participants in this study and the NIHR National BioResource. The views expressed are those of the author(s) and not necessarily those of the NIHR or the Department of Health and Social Care. JLG is funded by the Medical Research Council (MR/S010483/1; MR/P011705/1; MR/P01836X/1; MR/X012700/1). F.G., F.R., R.K. and A.L.G. were funded by the Wellcome Trust (grants 106262/Z/14/Z, 106263/Z/14/Z), the MRC Metabolic Diseases Unit (grants MRC MC UU 12012/3, MRC MC UU12012/5) and by the NIHR Cambridge Biomedical Research Centre. The mass spectrometers were obtained using the Medical Research Council "Enhancing UK Clinical Research" grant (MR/M009041/1). The views expressed are those of the author(s) and not necessarily those of the relevant funding or supporting institutions. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript
Collapse
Affiliation(s)
- Gabriele Mocciaro
- Roger Williams Institute of Liver StudiesFoundation for Liver ResearchLondonUK
| | - Amy L. George
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Michael Allison
- Liver Unit, Cambridge NIHR Biomedical Research CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Mattia Frontini
- Faculty of Health and Life Sciences, Clinical and Biomedical SciencesUniversity of Exeter Medical SchoolExeterUK
| | - Isabel Huang‐Doran
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Frank Reiman
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Fiona Gribble
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Julian L. Griffin
- The Rowett Institute, Foresterhill CampusUniversity of AberdeenAberdeenUK
| | - Antonio Vidal‐Puig
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Vian Azzu
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
- Liver Unit, Cambridge NIHR Biomedical Research CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Richard Kay
- Institute of Metabolic Science Metabolic Research LaboratoriesAddenbrooke's HospitalCambridgeUK
| | - Michele Vacca
- Roger Williams Institute of Liver StudiesFoundation for Liver ResearchLondonUK
- University of Bari "Aldo Moro"Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni"BariItaly
| |
Collapse
|
23
|
Schwenger KJP, Copeland JK, Ghorbani Y, Chen L, Comelli EM, Guttman DS, Fischer SE, Jackson TD, Okrainec A, Allard JP. Characterization of liver, adipose, and fecal microbiome in obese patients with MASLD: links with disease severity and metabolic dysfunction parameters. MICROBIOME 2025; 13:9. [PMID: 39810228 PMCID: PMC11730849 DOI: 10.1186/s40168-024-02004-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a range of histological findings from the generally benign simple steatosis to steatohepatitis (MASH) which can progress to fibrosis and cirrhosis. Several factors, including the microbiome, may contribute to disease progression. RESULTS Here, we demonstrate links between the presence and abundance of specific bacteria in the adipose and liver tissues, inflammatory genes, immune cell responses, and disease severity. Overall, in MASLD patients, we observed a generalized obesity-induced translocation of gut bacteria to hepatic and adipose tissues. We identified microbial patterns unique to more severely diseased tissues. Specifically, Enterococcus, Granulicatella, and Morganellaceae abundance is positively correlated with immune cell counts and inflammatory gene expression levels, and both genera are significantly enriched in MASH patients. Brevibacterium is enriched in adipose tissues of patients with liver fibrosis. CONCLUSION Together, these results provide further insight into the microbial factors that may be driving disease severity. Video Abstract.
Collapse
Affiliation(s)
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Canada
| | - Yasaman Ghorbani
- Toronto General Hospital, University Health Network, Toronto, Canada
| | - Lina Chen
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Canada
| | - Sandra E Fischer
- Toronto General Hospital, University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Timothy D Jackson
- Division of General Surgery, University of Toronto, Toronto, Canada
- Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Allan Okrainec
- Division of General Surgery, University of Toronto, Toronto, Canada
- Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada.
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada.
- Division of Gastroenterology, Department of Medicine, Toronto General Hospital, 585 University Avenue, 9N-973, Toronto, ON, M5G 2N2, Canada.
| |
Collapse
|
24
|
Gillet R, Cerda-Drago TG, Brañes MC, Valenzuela R. Submicron Dispersions of Phytosterols Reverse Liver Steatosis with Higher Efficacy than Phytosterol Esters in a Diet Induced-Fatty Liver Murine Model. Int J Mol Sci 2025; 26:564. [PMID: 39859279 PMCID: PMC11766071 DOI: 10.3390/ijms26020564] [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: 12/06/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Consumption of phytosterols is a nutritional strategy employed to reduce cholesterol absorption, but recent research shows that their biological activity might go beyond cholesterol reduction for the treatment of metabolic dysfunction-associated fatty liver disease (MAFLD), and novel phytosterol formulations, such as submicron dispersions, could improve these effects. We explored the therapeutic activity of phytosterols, either formulated as submicron dispersions of phytosterols (SDPs) or conventional phytosterol esters (PEs), in a mouse model of MAFLD. MAFLD was induced in mice by atherogenic diet (AD) feeding. The reversion of distorted serum and liver parameter values after a period of AD feeding was investigated after supplementation of the AD with SDPs, PEs, or a placebo (PT). Additionally, the metabolic parameters of fatty acid synthesis, fatty acid oxidation, and inflammation were studied to understand the mechanism of action of phytosterols. AD supplementation with SDPs was shown to reduce liver fat, along with showing a significant improvement in liver triglycerides (TGs), free fatty acids (FFAs), and liver cholesterol levels. These results were reinforced by the analyses of the liver steatosis scores, and liver histologies, where SDP intervention showed a consistent improvement. Treatment with PEs showed slighter effects in the same analyses, and no effects were observed with the PT treatment. Additionally, SDP intervention reversed, with a higher efficacy than PEs, the effect of AD on the serum levels of TGs, total- and LDL-cholesterol levels, and glucose levels. And, exceptionally, while SDP improved HDL-cholesterol serum levels, PEs did not show any effect on this parameter. We provide evidence for the therapeutical activity of phytosterols in MAFLD beyond the regulation of cholesterol levels, which is increased when the phytosterols are formulated as submicron dispersions compared to ester formulations.
Collapse
Affiliation(s)
- Raimundo Gillet
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - Tomás G. Cerda-Drago
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - María C. Brañes
- Naturalis Research Consortium, Santiago 8700548, Chile; (R.G.); (T.G.C.-D.); (M.C.B.)
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago 8380000, Chile
| |
Collapse
|
25
|
Gurjar S, Bhat A R, Upadhya R, Shenoy RP. Extracellular vesicle-mediated approaches for the diagnosis and therapy of MASLD: current advances and future prospective. Lipids Health Dis 2025; 24:5. [PMID: 39773634 PMCID: PMC11705780 DOI: 10.1186/s12944-024-02396-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an asymptomatic, multifaceted condition often associated with various risk factors, including fatigue, obesity, insulin resistance, metabolic syndrome, and sleep apnea. The increasing burden of MASLD underscores the critical need for early diagnosis and effective therapies. Owing to the lack of efficient therapies for MASLD, early diagnosis is crucial. Consequently, noninvasive biomarkers and imaging techniques are essential for analyzing disease risk and play a pivotal role in the global diagnostic process. The use of extracellular vesicles has emerged as promising for early diagnosis and therapy of various liver ailments. Herein, a comprehensive summary of the current diagnostic modalities for MASLD is presented, highlighting their advantages and limitations while exploring the potential of extracellular vesicles (EVs) as innovative diagnostic and therapeutic tools for MASLD. With this aim, this review emphasizes an in-depth understanding of the origin of EVs and the pathophysiological alterations of these ectosomes and exosomes in various liver diseases. This review also explores the therapeutic potential of EVs as key components in the future management of liver disease. The dual role of EVs as biomarkers and their therapeutic utility in MASLD essentially highlights their clinical integration to improve MASLD diagnosis and treatment. While EV-based therapies are still in their early stages of development and require substantial research to increase their therapeutic value before they can be used clinically, the diagnostic application of EVs has been extensively explored. Moving forward, developing diagnostic devices leveraging EVs will be crucial in advancing MASLD diagnosis. Thus, the literature summarized provides suitable grounds for clinicians and researchers to explore EVs for devising diagnostic and treatment strategies for MASLD.
Collapse
Affiliation(s)
- Swasthika Gurjar
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India
| | - Ramanarayana Bhat A
- Manipal Centre for Biotherapeutics Research, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India
| | - Raghavendra Upadhya
- Manipal Centre for Biotherapeutics Research, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India.
| | - Revathi P Shenoy
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Karnataka, 576104, Manipal, India.
| |
Collapse
|
26
|
Leopold M, Mass-Sanchez PB, Krizanac M, Štancl P, Karlić R, Prabutzki P, Parafianczuk V, Schiller J, Asimakopoulos A, Engel KM, Weiskirchen R. How the liver transcriptome and lipid composition influence the progression of nonalcoholic fatty liver disease to hepatocellular carcinoma in a murine model. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159574. [PMID: 39510374 DOI: 10.1016/j.bbalip.2024.159574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024]
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) has been steadily increasing in Western society in recent years and has been recognized as a risk factor for the development of hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying the progression from NAFLD to HCC are still unclear, despite the use of suitable mouse models. To identify the transcriptional and lipid profiles of livers from mice with NAFLD-HCC, we induced both NAFLD and NAFLD-HCC pathologies in C57BL/6J mice and performed RNA-sequencing (RNA-seq) and targeted lipidomic analysis. Our RNA-seq analysis revealed that the transcriptional signature of NAFLD in mice is characterized by changes in inflammatory response and fatty acid metabolism. Moreover, the signature of NAFLD-HCC is characterized by processes typically observed in cancer, such as epithelial to mesenchymal transition, angiogenesis and inflammatory responses. Furthermore, we found that the diet used in this study inhibited cholesterol synthesis in both models. The analysis of lipid composition also showed a significant impact of the provided diet. Therefore, our study supports the idea that a Western diet (WD) affects metabolic processes and hepatic lipid composition. Additionally, the combination of a WD with the administration of a carcinogen drives the progression from NAFLD to HCC.
Collapse
Affiliation(s)
- Marvin Leopold
- Institute for Medical Physics and Biophysics, Leipzig University, Faculty of Medicine, 04107 Leipzig, Germany; Klinik für Neurologie, Sana Klinikum Borna, 04552 Borna, Germany.
| | - Paola Berenice Mass-Sanchez
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany.
| | - Marinela Krizanac
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany.
| | - Paula Štancl
- Bioinformatics Group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia.
| | - Rosa Karlić
- Bioinformatics Group, Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia.
| | - Patricia Prabutzki
- Institute for Medical Physics and Biophysics, Leipzig University, Faculty of Medicine, 04107 Leipzig, Germany.
| | - Victoria Parafianczuk
- Institute for Medical Physics and Biophysics, Leipzig University, Faculty of Medicine, 04107 Leipzig, Germany
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, Leipzig University, Faculty of Medicine, 04107 Leipzig, Germany.
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany
| | - Kathrin M Engel
- Institute for Medical Physics and Biophysics, Leipzig University, Faculty of Medicine, 04107 Leipzig, Germany.
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany.
| |
Collapse
|
27
|
Kobayashi T, Nakatsuka T, Sato M, Soroida Y, Hikita H, Gotoh H, Iwai T, Tateishi R, Kurano M, Fujishiro M. Diagnostic performance of two-dimensional shear wave elastography and attenuation imaging for fibrosis and steatosis assessment in chronic liver disease. J Med Ultrason (2001) 2025; 52:95-103. [PMID: 38951430 PMCID: PMC11799025 DOI: 10.1007/s10396-024-01473-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/14/2024] [Indexed: 07/03/2024]
Abstract
PURPOSE We investigated the diagnostic performance of two-dimensional shear wave elastography (2D-SWE) and attenuation imaging (ATI) in detecting fibrosis and steatosis in patients with chronic liver disease (CLD), comparing them with established methods. METHODS In 190 patients with CLD, 2D-SWE and vibration-controlled transient elastography (VCTE) were used for liver stiffness measurement (LSM), and ATI and controlled attenuation parameter (CAP) were used for steatosis quantification. The correlations between these new and established methods were analyzed. RESULTS Significant correlations were found between 2D-SWE and VCTE (r = 0.78, P < 0.001), and between ATI and CAP (r = 0.70, P < 0.001). Liver stiffness tended to be lower with 2D-SWE compared with that with VCTE, especially in cases with higher LSM, and ATI was less influenced by skin-capsular distance than CAP. Area under the receiver-operating characteristics curves (AUCs) and optimal cut-offs of 2D-SWE for diagnosing liver fibrosis stages F2, F3, and F4 were 0.73 (8.7 kPa), 0.79 (9.1 kPa), and 0.88 (11.6 kPa), respectively. The AUCs and optimal cut-offs of ATI for diagnosing hepatic steatosis grades S1, S2, and S3 were 0.91 (0.66 dB/cm/MHz), 0.80 (0.79 dB/cm/MHz), and 0.88 (0.86 dB/cm/MHz), respectively. A subgroup analysis of 86 patients with metabolic dysfunction-associated steatotic liver disease also demonstrated good performance for 2D-SWE and ATI. CONCLUSION 2D-SWE and ATI performed comparably with conventional VCTE and CAP in evaluating CLD, offering reliable alternatives for diagnosing liver fibrosis and steatosis.
Collapse
Affiliation(s)
- Tamaki Kobayashi
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| | - Masaya Sato
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Yoko Soroida
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiromi Hikita
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Hiroaki Gotoh
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Tomomi Iwai
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| |
Collapse
|
28
|
Patel K, Asrani SK, Fiel MI, Levine D, Leung DH, Duarte-Rojo A, Dranoff JA, Nayfeh T, Hasan B, Taddei TH, Alsawaf Y, Saadi S, Majzoub AM, Manolopoulos A, Alzuabi M, Ding J, Sofiyeva N, Murad MH, Alsawas M, Rockey DC, Sterling RK. Accuracy of blood-based biomarkers for staging liver fibrosis in chronic liver disease: A systematic review supporting the AASLD Practice Guideline. Hepatology 2025; 81:358-379. [PMID: 38489517 DOI: 10.1097/hep.0000000000000842] [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: 11/08/2023] [Accepted: 02/19/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND AND AIMS Blood-based biomarkers have been proposed as an alternative to liver biopsy for noninvasive liver disease assessment in chronic liver disease. Our aims for this systematic review were to evaluate the diagnostic utility of selected blood-based tests either alone, or in combination, for identifying significant fibrosis (F2-4), advanced fibrosis (F3-4), and cirrhosis (F4), as compared to biopsy in chronic liver disease. APPROACH AND RESULTS We included a comprehensive search of databases including Ovid MEDLINE(R), EMBASE, Cochrane Database, and Scopus through to April 2022. Two independent reviewers selected 286 studies with 103,162 patients. The most frequently identified studies included the simple aspartate aminotransferase-to-platelet ratio index and fibrosis (FIB)-4 markers (with low-to-moderate risk of bias) in HBV and HCV, HIV-HCV/HBV coinfection, and NAFLD. Positive (LR+) and negative (LR-) likelihood ratios across direct and indirect biomarker tests for HCV and HBV for F2-4, F3-4, or F4 were 1.66-6.25 and 0.23-0.80, 1.89-5.24 and 0.12-0.64, and 1.32-7.15 and 0.15-0.86, respectively; LR+ and LR- for NAFLD F2-4, F3-4, and F4 were 2.65-3.37 and 0.37-0.39, 2.25-6.76 and 0.07-0.87, and 3.90 and 0.15, respectively. Overall, the proportional odds ratio indicated FIB-4 <1.45 was better than aspartate aminotransferase-to-platelet ratio index <0.5 for F2-4. FIB-4 >3.25 was also better than aspartate aminotransferase-to-platelet ratio index >1.5 for F3-4 and F4. There was limited data for combined tests. CONCLUSIONS Blood-based biomarkers are associated with small-to-moderate change in pretest probability for diagnosing F2-4, F3-4, and F4 in viral hepatitis, HIV-HCV coinfection, and NAFLD, with limited comparative or combination studies for other chronic liver diseases.
Collapse
Affiliation(s)
- Keyur Patel
- Department of Medcine, Division of Gastroenterology and Hepatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Sumeet K Asrani
- Department of Medicine, Division of Hepatology, Baylor University Medical Center, Dallas, Texas, USA
| | - Maria Isabel Fiel
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Deborah Levine
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel H Leung
- Department of Pediatrics, Baylor College of Medicine and Division of Gastroenterology, Hepatology and Nutrition, Texas Children's Hospital, Houston, Texas, USA
| | - Andres Duarte-Rojo
- Division of Gastroenterology and Hepatology, Northwestern Medicine and Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jonathan A Dranoff
- Yale School of Medicine, Department of Internal Medicine, Section of Digestive Diseases, New Haven, Connecticut, USA
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Tarek Nayfeh
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Bashar Hasan
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamar H Taddei
- Yale School of Medicine, Department of Internal Medicine, Section of Digestive Diseases, New Haven, Connecticut, USA
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Yahya Alsawaf
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Samer Saadi
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Muayad Alzuabi
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Jingyi Ding
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Nigar Sofiyeva
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Mohammad H Murad
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Mouaz Alsawas
- Mayo Clinic Evidence-based Practice Center, Mayo Clinic, Rochester, Minnesota, USA
- Department of Medicine, Section of Hepatology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Don C Rockey
- Department of Medicine, Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Richard K Sterling
- Department of Medicine, Section of Hepatology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
29
|
Flam E, Haas JT, Staels B. Liver metabolism in human MASLD: A review of recent advancements using human tissue metabolomics. Atherosclerosis 2025; 400:119054. [PMID: 39586140 DOI: 10.1016/j.atherosclerosis.2024.119054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/27/2024]
Abstract
Global incidence of Metabolic dysfunction-Associated Steatotic Liver Disease (MASLD) is on the rise while treatments remain elusive. MASLD is a disease of dysregulated systemic and hepatic metabolism. Current understanding of disease pathophysiology as it relates to metabolome changes largely comes from studies on animal models and human plasma. However, human tissue data are crucial for transitioning from mechanisms to clinical therapies. The close relationship between MASLD and comorbidities like obesity, type 2 diabetes and dyslipidemia make it difficult to determine the contribution from liver disease itself. Here, we review recent metabolomics studies in liver tissue from human MASLD patients, which have predominately focused on lipid metabolism, but also include bile acid, tricarboxylic acid (TCA) cycle, and branched chain amino acid (BCAA) metabolism. Several clinical trials are underway to target various of these lipid-related pathways in MASLD. Although only the β-selective thyroid hormone receptor agonist resmetirom has so far been approved for use, many metabolism-targeting pharmaceuticals show promising results for halting disease progression, if not promoting outright reversal. Ultimately, the scarcity of human tissue data and the variability of confounding factors, like obesity, within and between cohorts are impediments to the pathophysiological understanding required for efficient development of metabolic treatments.
Collapse
Affiliation(s)
- Emily Flam
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
30
|
Pickhardt PJ, Lubner MG. Noninvasive Quantitative CT for Diffuse Liver Diseases: Steatosis, Iron Overload, and Fibrosis. Radiographics 2025; 45:e240176. [PMID: 39700040 DOI: 10.1148/rg.240176] [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: 12/21/2024]
Abstract
Chronic diffuse liver disease continues to increase in prevalence and represents a global health concern. Noninvasive detection and quantification of hepatic steatosis, iron overload, and fibrosis are critical, especially given the many relative disadvantages and potential risks of invasive liver biopsy. Although MRI techniques have emerged as the preferred reference standard for quantification of liver fat, iron, and fibrosis, CT can play an important role in opportunistic detection of unsuspected disease and is performed at much higher volumes. For hepatic steatosis, noncontrast CT provides a close approximation to MRI-based proton-density fat fraction (PDFF) quantification, with liver attenuation values less than or equal to 40 HU signifying at least moderate steatosis. Liver fat quantification with postcontrast CT is less precise but can generally provide categorical assessment (eg, mild vs moderate steatosis). Noncontrast CT can also trigger appropriate assessment for iron overload when increased parenchymal attenuation values are observed (eg, >75 HU). A variety of morphologic and functional CT features indicate the presence of underlying hepatic fibrosis and cirrhosis. Beyond subjective assessment, quantitative CT methods for staging fibrosis can provide comparable performance to that of elastography. Furthermore, quantitative CT assessment can be performed retrospectively, since prospective techniques are not required. Many of these CT quantitative measures are now fully automated via artificial intelligence (AI) deep learning algorithms. These retrospective and automated advantages have important implications for longitudinal clinical care and research. Ultimately, regardless of the indication for CT, opportunistic detection of steatosis, iron overload, and fibrosis can result in appropriate clinical awareness and management. ©RSNA, 2024 See the invited commentary by Yeh in this issue.
Collapse
Affiliation(s)
- Perry J Pickhardt
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, E3/311 Clinical Science Center, Madison, WI 53792-3252; and the American College of Radiology (ACR) Institute for Radiologic Pathology, Silver Spring, Md
| | - Meghan G Lubner
- From the Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, E3/311 Clinical Science Center, Madison, WI 53792-3252; and the American College of Radiology (ACR) Institute for Radiologic Pathology, Silver Spring, Md
| |
Collapse
|
31
|
Qi H, Jiang S, Nan J, Guo H, Cheng C, He X, Jin H, Zhang R, Lei J. Application and research progress of magnetic resonance proton density fat fraction in metabolic dysfunction-associated steatotic liver disease: a comprehensive review. Abdom Radiol (NY) 2025; 50:185-197. [PMID: 39048719 DOI: 10.1007/s00261-024-04448-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/27/2024]
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), formerly known as Non-Alcoholic Fatty Liver Disease (NAFLD), is a chronic liver disorder associated with disturbances in lipid metabolism. The disease is prevalent worldwide, particularly closely linked with metabolic syndromes such as obesity and diabetes. Magnetic Resonance Proton Density Fat Fraction (MRI-PDFF), serving as a non-invasive and highly quantitative imaging assessment tool, holds promising applications in the diagnosis and research of MASLD. This paper aims to comprehensively review and summarize the applications and research progress of MRI-PDFF technology in MASLD, analyze its strengths and challenges, and anticipate its future developments in clinical practice.
Collapse
Affiliation(s)
- Hongyan Qi
- The First Clinical Medical College of Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou City, 730000, Gansu Province, China
| | | | - Jiang Nan
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hang Guo
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Cai Cheng
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xin He
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hongyang Jin
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Rongfan Zhang
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Junqiang Lei
- The First Clinical Medical College of Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou City, 730000, Gansu Province, China.
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
- Radiological Clinical Medicine Research Center of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
32
|
Nakamura Y, Hirooka M, Koizumi Y, Yano R, Imai Y, Watanabe T, Yoshida O, Tokumoto Y, Abe M, Hiasa Y. Diagnostic accuracy of ultrasound-derived fat fraction for the detection and quantification of hepatic steatosis in patients with liver biopsy. J Med Ultrason (2001) 2025; 52:85-94. [PMID: 38918301 DOI: 10.1007/s10396-024-01472-6] [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: 01/05/2024] [Accepted: 05/13/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE This retrospective study was conducted to investigate the diagnostic accuracy of ultrasound-derived fat fraction (UDFF) for grading hepatic steatosis using liver histology as the reference standard. METHODS Seventy-three patients with liver disease were assessed using UDFF and liver biopsy. Pearson's test and the Bland-Altman plot were used to assess the correlation between UDFF and histological fat content in liver sections. The UDFF cutoff values for histologically proven steatosis grades were determined using the area under the receiver operating characteristic curve (AUROC). RESULTS The median age of the patients was 66 (interquartile range 54-74) years, and 33 (45%) were females. The UDFF values showed a stepwise increase with increasing steatosis grade (p < .001) and were strongly correlated with the histological fat content (r = .7736, p < .001). The Bland-Altman plot revealed a mean bias of 2.384% (95% limit of agreement, - 6.582 to 11.351%) between them. Univariate regression analysis revealed no significant predictors of divergence. The AUROCs for distinguishing steatosis grades of ≥ 1, ≥2, and 3 were 0.956 (95% confidence interval [CI], 0.910-1.00), 0.926 (95% CI, 0.860-0.993), and 0.971 (95% CI, 0.929-1.000), respectively. The UDFF cutoff value of > 6% had a sensitivity and specificity of 94.8% and 82.3%, respectively, for diagnosing steatosis grade ≥ 1. There was no association between UDFF and the fibrosis stage. CONCLUSION UDFF shows strong agreement with the histological fat content and excellent diagnostic accuracy for grading steatosis. UDFF is a promising tool for detecting and quantifying hepatic steatosis in clinical practice.
Collapse
Affiliation(s)
- Yoshiko Nakamura
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan.
| | - Yohei Koizumi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Ryo Yano
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Yusuke Imai
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Takao Watanabe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Yoshio Tokumoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Masanori Abe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, 454 Toon, Ehime, 791-0295, Japan
| |
Collapse
|
33
|
Ishiba H, Fujii H, Kamada Y, Sumida Y, Takahashi H, Seko Y, Toyoda H, Hayashi H, Yamaguchi K, Iwaki M, Yoneda M, Arai T, Shima T, Morishita A, Kawata K, Tomita K, Kawanaka M, Yoshida Y, Ikegami T, Notsumata K, Oeda S, Fukushima H, Miyoshi E, Aishima S, Itoh Y, Okanoue T, Nakajima A. Accuracy of type IV collagen 7S versus Enhanced Liver Fibrosis score for diagnosing fibrosis in patients with metabolic dysfunction-associated steatotic liver disease. Hepatol Commun 2025; 9:e0563. [PMID: 39670882 PMCID: PMC11637746 DOI: 10.1097/hc9.0000000000000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Various noninvasive tests can be used to identify high-risk groups of patients with metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD). In this study, we compared the diagnostic performance of serum type 4 collagen 7S (COL4-7S) and the Enhanced Liver Fibrosis (ELF) score for detecting fibrosis in patients with MASLD. METHODS Among 1368 patients with MASLD who underwent liver biopsy, 794 with values for both serum COL4-7S and the ELF score were enrolled in this multicenter study. The diagnostic performance of COL4-7S and ELF for detecting fibrosis stage ≥2, fibrosis stage ≥3, and at-risk metabolic dysfunction-associated steatohepatitis were evaluated using ROC curve, continuous net reclassification improvement, and integrated discrimination improvement analyses. RESULTS Both COL4-7S and ELF scores increased significantly with increasing fibrosis. The AUROC for each outcome was higher for COL4-7S than ELF, but not significantly. The diagnostic performance for detecting fibrosis stage ≥2 was significantly better for COL4-7S than for the ELF score (s net reclassification improvement=16.7%, p=0.018; integrated discrimination improvement=3.9%, p<0.01). In patients without diabetes, the diagnostic performance for each outcome did not differ significantly between COL4-7S and ELF score, but in patients with diabetes, the diagnostic performance for fibrosis stage ≥2 was higher for COL4-7S than for the ELF score (AUROC=0.817 vs. 0.773, p=0.04; s net reclassification improvement=32.7%, p<0.01; integrated discrimination improvement=5.6%, p<0.01). CONCLUSIONS The diagnostic performance of serum COL4-7S (a single marker) for identifying more advanced disease in patients with MASLD was at least equivalent to that of the ELF score (a combined marker).
Collapse
Affiliation(s)
- Hiroshi Ishiba
- Department of Gastroenterology, Osaka General Hospital of West Japan Railway Company, Abeno, Osaka, Japan
| | - Hideki Fujii
- Department of Hepatology, Osaka Metropolitan University, Osaka, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita,Japan
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare University, Tokyo, Japan
| | | | - Yuya Seko
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Hideki Hayashi
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
| | - Kanji Yamaguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | - Taeang Arai
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Toshihide Shima
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kazuhito Kawata
- Department of Internal Medicine II, Hepatology Division, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kengo Tomita
- Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, General Medical Center, Kawasaki Medical School, Kitaku, Okayama, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Kishibeshinmachi, Osaka, Japan
| | - Tadashi Ikegami
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Kazuo Notsumata
- Department of General Internal Medicine, Fukui-ken Saiseikai Hospital, 7-1 Wadanaka-chou Funabashi, Fukui, Japan
| | - Satoshi Oeda
- Liver Center, Saga University Hospital, Saga, Japan
| | - Hideaki Fukushima
- Diagnostics Business Area, Siemens Healthcare Diagnostics K.K., Osaki Shinagawa-ku, Tokyo, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry & Clinical Investigation, Osaka University Graduate School of Medicine, Yamada-oka, Suita, Osaka, Japan
| | - Shinichi Aishima
- Department of Scientific Pathology Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| |
Collapse
|
34
|
Yang Z, Chen Q, Wang J, Qiu Y, Thepsuwan P, Yi Z, Heng HH, Sun Q, Chen X, Li L, He P, Zhang R, Zhang K. Inhalation exposure to airborne PM 2.5 attenuates hepatic metabolic pathways through S-nitrosylation of the primary ER stress sensor. Am J Physiol Cell Physiol 2025; 328:C212-C226. [PMID: 39607384 PMCID: PMC11901345 DOI: 10.1152/ajpcell.00385.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Inhalation exposure to airborne fine particulate matter (aerodynamic diameter: <2.5 µm, PM2.5) is known to cause metabolic dysfunction-associated steatohepatitis (MASH) and the associated metabolic syndrome. Hepatic lipid accumulation and inflammation are the key characteristics of MASH. However, the mechanism by which PM2.5 exposure induces lipid accumulation and inflammation in the liver remains to be further elucidated. In this study, we revealed that inhalation exposure to PM2.5 induces nitrosative stress in mouse livers by suppressing hepatic S-nitrosoglutathione reductase activities, which leads to S-nitrosylation modification of the primary unfolded protein response (UPR) transducer inositol-requiring 1 α (IRE1α), an endoplasmic reticulum-resident protein kinase and endoribonuclease (RNase). S-nitrosylation suppresses the RNase activity of IRE1α and subsequently decreases IRE1α-mediated splicing of the mRNA encoding X-box binding protein 1 (XBP1) and IRE1α-dependent degradation of select microRNAs (miRNAs), including miR-200 family members, miR-34, miR-223, miR-155, and miR-146, in the livers of the mice exposed to PM2.5. Elevation of IRE1α-target miRNAs, due to impaired IRE1α RNase activity by PM2.5-triggered S-nitrosylation, leads to decreased expression of the major regulators of fatty acid oxidation, lipolysis, and anti-inflammatory response, including XBP1, sirtuin 1, peroxisome proliferator-activated receptor α, and peroxisome proliferator-activated receptor γ, in the liver, which account at least partially for hepatic lipid accumulation and inflammation in mice exposed to airborne PM2.5. In summary, our study revealed a novel pathway by which PM2.5 causes cytotoxicity and promotes MASH-like phenotypes through inducing hepatic nitrosative stress and S-nitrosylation of the primary UPR transducer and subsequent elevation of select miRNAs involved in metabolism and inflammation in the liver.NEW & NOTEWORTHY Exposure to fine airborne particulate matter PM2.5 causes metabolic dysfunction-associated steatohepatitis characterized by hepatic steatosis, inflammation, and fibrosis. Here, we discovered that inhalation exposure to environmental PM2.5 induces nitrosative stress in livers by suppressing hepatic S-nitrosoglutathione reductase activities, which leads to S-nitrosylation of the unfolded protein response transducer IRE1α. S-nitrosylation decreases IRE1α-dependent degradation of miRNAs in the livers of mice exposed to PM2.5, leading to downregulation of major regulators of energy metabolism and anti-inflammatory response.
Collapse
Affiliation(s)
- Zhao Yang
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Qi Chen
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Jiemei Wang
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, Michigan, United States
| | - Yining Qiu
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Pattaraporn Thepsuwan
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Zhengping Yi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, Michigan, United States
| | - Henry H Heng
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Qinghua Sun
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Xuequn Chen
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Li Li
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Peijian He
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Ren Zhang
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Kezhong Zhang
- Center for Molecular Medicine & Genetics, The Wayne State University School of Medicine, Detroit, Michigan, United States
- Department of Biochemistry, Microbiology, and Immunology, The Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
35
|
Hirata Y, Sakuma Y, Ogiso H, Nagai R, Aizawa K. Targeted Plasma Bile Acid Metabolomic Analysis in Metabolic Dysfunction-Associated Steatohepatitis and Alcoholic Hepatitis. Biomedicines 2024; 13:78. [PMID: 39857662 PMCID: PMC11762544 DOI: 10.3390/biomedicines13010078] [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: 12/05/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Even though many metabolic liver diseases can now be diagnosed using blood tests and diagnostic imaging, early diagnosis remains difficult. Understanding mechanisms contributing to the progression from Metabolic Dysfunction-Associated Steatohepatitis (MASH) and Alcoholic Hepatitis (AH) to cirrhosis is critical to reduce the burden of end-stage liver disease. Monitoring individual bile acids has been proposed as a way to distinguish various liver disorders. Methods: This study explored bile acid profiles in patients with MASH and AH. Plasma samples from patients with MASH, AH, and a control group were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) to quantify bile acid concentrations. Targeted metabolomic analysis was performed to compare bile acid levels between the hepatitis and control groups. Results: Concentrations of ursodeoxycholic acid (UDCA), chenodeoxycholic acid (CDCA), taurocholic acid (TCA), tauroursodeoxycholic acid (TUDCA), taurochenodeoxycholic acid (TCDCA), glycoursodeoxycholic acid (GUDCA), glycochenodeoxycholic acid (GCDCA), and glycocholic acid (GCA) were significantly elevated in the hepatitis group. Correlation analysis revealed strong positive relationships between the total and direct bilirubin levels and TUDCA and GCDCA. Aspartate aminotransferase (AST) showed strong positive correlations with TCDCA and GCDCA. Child-Pugh score, Fibrosis-4 index, and non-alcoholic fatty liver disease fibrosis score were positively correlated with GCA, whereas the aspartate aminotransferase-to-platelet ratio correlated with TCA, TCDCA, and GCA. The model for end-stage liver disease (MELD) score showed a strong positive correlation with GCDCA. Implications: GCDCA may serve as a predictive biomarker for liver damage, potentially enabling early diagnosis and targeted intervention in patients with MASH and AH.
Collapse
Affiliation(s)
- Yuta Hirata
- Division of Gastroenterological, Department of Surgery, General and Transplant Surgery, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan
| | - Yasunaru Sakuma
- Division of Gastroenterological, Department of Surgery, General and Transplant Surgery, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan
| | - Hideo Ogiso
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan
- Clinical Pharmacology Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Tochigi, Japan
- Division of Translational Research, Clinical Research Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Tochigi, Japan
| |
Collapse
|
36
|
Wang XX, Song YY, Jin R, Wang ZL, Li XH, Yang Q, Teng X, Liu FF, Wu N, Xie YD, Rao HY, Liu F. Hepatic Steatosis Analysis in Metabolic Dysfunction-Associated Steatotic Liver Disease Based on Artificial Intelligence. Diagnostics (Basel) 2024; 14:2889. [PMID: 39767250 PMCID: PMC11675354 DOI: 10.3390/diagnostics14242889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by the accumulation of fat in the liver, excluding excessive alcohol consumption and other known causes of liver injury. Animal models are often used to explore different pathogenic mechanisms and therapeutic targets of MASLD. The aim of this study is to apply an artificial intelligence (AI) system based on second-harmonic generation (SHG)/two-photon-excited fluorescence (TPEF) technology to automatically assess the dynamic patterns of hepatic steatosis in MASLD mouse models. METHODS We evaluated the characteristics of hepatic steatosis in mouse models of MASLD using AI analysis based on SHG/TPEF images. Six different models of MASLD were induced in C57BL/6 mice by feeding with a western or high-fat diet, with or without fructose in their drinking water, and/or by weekly injections of carbon tetrachloride. RESULTS Body weight, serum lipids, and liver enzyme markers increased at 8 and 16 weeks in each model compared to baseline. Steatosis grade showed a steady upward trend. However, the non-alcoholic steatohepatitis (NASH) Clinical Research Network (CRN) histological scoring method detected no significant difference between 8 and 16 weeks. In contrast, AI analysis was able to quantify dynamic changes in the area, number, and size of hepatic steatosis automatically and objectively, making it more suitable for preclinical MASLD animal experiments. CONCLUSIONS AI recognition technology may be a new tool for the accurate diagnosis of steatosis in MASLD, providing a more precise and objective method for evaluating steatosis in preclinical murine MASLD models under various experimental and treatment conditions.
Collapse
Affiliation(s)
- Xiao-Xiao Wang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Yu-Yun Song
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Rui Jin
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Zi-Long Wang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Xiao-He Li
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Qiang Yang
- Hangzhou Choutu Technology Co., Ltd., Hangzhou 310052, China;
| | - Xiao Teng
- HistoIndex Pte Ltd., Singapore 117674, Singapore;
| | - Fang-Fang Liu
- Department of Pathology, Peking University People’s Hospital, Beijing 100044, China;
| | - Nan Wu
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Yan-Di Xie
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Hui-Ying Rao
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| | - Feng Liu
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing 100044, China; (X.-X.W.); (Y.-Y.S.); (R.J.); (Z.-L.W.); (X.-H.L.); (N.W.); (Y.-D.X.); (H.-Y.R.)
| |
Collapse
|
37
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [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: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
38
|
Le Jan D, Siliman Misha M, Destrumelle S, Terceve O, Thorin C, Larcher T, Ledevin M, Desfontis JC, Betti E, Mallem Y. Omega-3 Fatty Acid and Vitamin D Supplementations Partially Reversed Metabolic Disorders and Restored Gut Microbiota in Obese Wistar Rats. BIOLOGY 2024; 13:1070. [PMID: 39765737 PMCID: PMC11673857 DOI: 10.3390/biology13121070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025]
Abstract
Obesity is a global public health issue linked to various comorbidities in both humans and animals. This study investigated the effects of vitamin D (VD) and omega-3 fatty acids (ω3FA) on obesity, gut dysbiosis, and metabolic alterations in Wistar rats. After 13 weeks on a standard (S) or High-Fat, High-Sugar (HFHS) diet, the rats received VD, ω3FA, a combination (VD/ω3), or a control (C) for another 13 weeks. The HFHS diet led to increased weight gain, abdominal circumference, glucose intolerance, insulin resistance, and gut dysbiosis. VD supplementation improved their fasting blood glucose and reduced liver damage, while ω3FA slowed BMI progression, reduced abdominal fat, liver damage, and intestinal permeability, and modulated the gut microbiota. The combination of VD/ω3 prevented weight gain, decreased abdominal circumference, improved glucose tolerance, and reduced triglycerides. This study demonstrates that VD and ω3FA, alone or combined, offer significant benefits in preventing obesity, gut dysbiosis, and metabolic alterations, with the VD/ω3 combination showing the most promise. Further research is needed to explore the mechanisms behind these effects and their long-term potential in both animal and human obesity management.
Collapse
Affiliation(s)
- Dylan Le Jan
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| | - Mohamed Siliman Misha
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| | - Sandrine Destrumelle
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| | - Olivia Terceve
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| | - Chantal Thorin
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Oniris, UMR 703, PanTher, APEX, 44307 Nantes, France; (C.T.); (T.L.); (M.L.)
| | - Thibaut Larcher
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Oniris, UMR 703, PanTher, APEX, 44307 Nantes, France; (C.T.); (T.L.); (M.L.)
| | - Mireille Ledevin
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Oniris, UMR 703, PanTher, APEX, 44307 Nantes, France; (C.T.); (T.L.); (M.L.)
| | - Jean-Claude Desfontis
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| | - Eric Betti
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| | - Yassine Mallem
- Nutrition, PathoPhysiology and Pharmacology (NP3) Unit, Oniris, 101 Rte de Gachet, 44300 Nantes, France; (M.S.M.); (S.D.); (O.T.); (J.-C.D.); (E.B.)
| |
Collapse
|
39
|
Ratziu V. Cirrhose métabolique : une entité en plein essor. BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2024. [DOI: 10.1016/j.banm.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Williams DM, Ali J, Cragg J, Ch'ng CL, Williams NW, Stephens JW, Min T. The Bidirectional Relationship Between Type 2 Diabetes and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Retrospective Cohort Study. Cureus 2024; 16:e75993. [PMID: 39835079 PMCID: PMC11743228 DOI: 10.7759/cureus.75993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Type 2 diabetes (T2D) and metabolic dysfunction-associated steatotic liver disease (MASLD) have shared pathophysiology. We aim to explore associations between these diseases and the impact of T2D therapies on MASLD-related outcomes in a real-world population. Methods A retrospective cohort study included 153 patients with biopsy-proven MASLD. Health records were reviewed for biochemical or radiological changes over follow-up and compared by T2D status. The rate of incident T2D was determined, and in those with T2D, the changes over follow-up were compared by prescribed treatment. The statistical significance of changes over follow-up was evaluated by Student's t-test, and logistic regression was undertaken to determine the impact of variables on T2D development. Results One hundred and fifty-three patients were included with a mean follow-up of 48.0±22.0 months. Patients with T2D (n=73) were older than patients without T2D (n=80; 56.3 vs 51.9 years, p<0.05). Patients with T2D had a greater stage of hepatic fibrosis (2.6 vs 1.7, p<0.001). Nine (12.3%) patients with T2D and four (5.0%) without T2D died during follow-up (p=0.10). Patients without T2D had greater glycosylated haemoglobin (HbA1c) over follow-up (3.0 mmol/mol, p<0.01), and 21 (26.3%) developed T2D. Patients with T2D treated with sodium-glucose transporter-2 inhibitors (SGLT-2i) and/or glucagon-like peptide-1 receptor analogues (GLP-1RA) had a reduction in FibroScan®-controlled attenuation parameter (-33.7dB/m, p<0.001) but not liver stiffness measure. There were no significant FibroScan® changes in those receiving other treatments. Conclusions Patients with T2D had greater hepatic fibrosis, and one in four patients with MASLD developed T2D over four years. Treatment with SGLT-2i and/or GLP-1RA in patients with T2D is associated with improved measures of steatosis but not fibrosis.
Collapse
Affiliation(s)
- David M Williams
- Department of Diabetes and Endocrinology, Morriston Hospital, Swansea, GBR
| | - Jumaina Ali
- Department of General Medicine, Morriston Hospital, Swansea, GBR
| | - Jake Cragg
- Department of General Medicine, Morriston Hospital, Swansea, GBR
| | - Chin L Ch'ng
- Department of Hepatology, Singleton Hospital, Swansea, GBR
| | | | - Jeffrey W Stephens
- Diabetes Research Group, Swansea University Medical School, Swansea, GBR
| | - Thinzar Min
- Diabetes Research Group, Swansea University Medical School, Swansea, GBR
| |
Collapse
|
41
|
Tamura Y, Naganuma A, Suzuki Y, Uehara S, Hoshino T, Hatanaka T, Shibusawa N, Uehara A, Ogawa A, Kakizaki S, Uraoka T. Drug-induced Steatohepatitis Caused by Long-term Use of Topical Steroids for Atopic Dermatitis. Intern Med 2024; 63:3165-3170. [PMID: 38599864 PMCID: PMC11671201 DOI: 10.2169/internalmedicine.3439-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/19/2024] [Indexed: 04/12/2024] Open
Abstract
Atopic dermatitis is common in children and often treated with topical corticosteroids (TCs). A boy in his late teens who had been using TCs for atopic dermatitis was diagnosed with liver damage during a health checkup. A medical examination revealed severe steatotic liver disease and elevated liver enzyme levels despite the absence of typical symptoms such as central obesity. After discontinuation of TCs, an improvement in liver enzyme levels was observed, leading to the diagnosis of drug-induced steatohepatitis. This case underscores the potential liver risks associated with prolonged TC use in children, highlighting the need for parental education.
Collapse
Affiliation(s)
- Yuki Tamura
- Department of Gastroenterology, NHO Takasaki General Medical Center, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, NHO Takasaki General Medical Center, Japan
| | - Yuhei Suzuki
- Department of Gastroenterology, NHO Takasaki General Medical Center, Japan
| | - Sanae Uehara
- Department of Gastroenterology, NHO Takasaki General Medical Center, Japan
| | - Takashi Hoshino
- Department of Gastroenterology, NHO Takasaki General Medical Center, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Japan
| | - Nobuyuki Shibusawa
- Department of Endocrinology and Metabolism, NHO Takasaki General Medical Center, Japan
| | - Akihito Uehara
- Department of Dermatology, NHO Takasaki General Medical Center, Japan
| | - Akira Ogawa
- Department of Pathology, NHO Takasaki General Medical Center, Japan
| | - Satoru Kakizaki
- Department of Gastroenterology, NHO Takasaki General Medical Center, Japan
- Department of Clinical Research, NHO Takasaki General Medical Center, Japan
| | - Toshio Uraoka
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Japan
| |
Collapse
|
42
|
Babu AF, Palomurto S, Kärjä V, Käkelä P, Lehtonen M, Hanhineva K, Pihlajamäki J, Männistö V. Metabolic signatures of metabolic dysfunction-associated steatotic liver disease in severely obese patients. Dig Liver Dis 2024; 56:2103-2110. [PMID: 38825414 DOI: 10.1016/j.dld.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024]
Abstract
BACKROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) can lead to liver fibrosis, cirrhosis, and hepatocellular carcinoma. Still, most patients with MASLD die from cardiovascular diseases indicating metabolic alterations related to both liver and cardiovascular pathology. AIMS AND METHODS The aim of this study was to assess biologic pathways behind MASLD progression from steatosis to metabolic dysfunction-associated steatohepatitis (MASH) using non-targeted liquid chromatography-mass spectrometry analysis in 106 severely obese individuals (78 women, mean age 47.7 7 ± 9.2 years, body mass index 41.8 ± 4.3 kg/m²) undergoing laparoscopic Roux-en-Y gastric bypass. RESULTS We identified several metabolites that are associated with MASLD progression. Most importantly, we observed a decrease of lysophosphatidylcholines LPC(18:2), LPC(18:3), and LPC(20:3) and increase of xanthine when comparing those with steatosis to those with MASH. We found that indole propionic acid and threonine were negatively correlated to fibrosis, but not with the metabolic disturbances associated with cardiovascular risk. Xanthine, ketoleucine, and tryptophan were positively correlated to lobular inflammation and ballooning but also with insulin resistance, and dyslipidemia, respectively. The results did not change when taking into account the most important genetic risk factors of MASLD. CONCLUSIONS Our findings suggest that there are several separate biological pathways, some of them independent of insulin resistance and dyslipidemia, associating with MASLD.
Collapse
Affiliation(s)
- Ambrin Farizah Babu
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland
| | - Saana Palomurto
- Department of Surgery, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Pirjo Käkelä
- Department of Surgery, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, Faculty of Health Science, University of Eastern Finland, 70211 Kuopio, Finland; LC-MS Metabolomics Center, Biocenter Kuopio, 70211 Kuopio, Finland
| | - Kati Hanhineva
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, 20014 Turku, Finland
| | - Jussi Pihlajamäki
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70210 Kuopio Finland
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland.
| |
Collapse
|
43
|
Schwenger KJP, Ghorbani Y, Bharatselvam S, Chen L, Chomiak KM, Tyler AC, Eddingsaas NC, Fischer SE, Jackson TD, Okrainec A, Allard JP. Links between fecal microplastics and parameters related to metabolic dysfunction-associated steatotic liver disease (MASLD) in humans: An exploratory study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:176153. [PMID: 39260480 DOI: 10.1016/j.scitotenv.2024.176153] [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: 07/17/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Microplastics (MPs) can persist in the environment and human body. Murine studies showed that exposure to MPs could cause metabolic dysregulation, contributing metabolic dysfunction-associated steatotic liver disease (MASLD) or steatohepatitis (MASH). However, research on the role of MPs in humans is limited. Thus, we aimed to assess links between human fecal MPs and liver histology, gene expression, immune cells and intestinal microbiota (IM). We included 6 lean healthy liver donors and 6 normal liver (obese) and 11 MASH patients. Overall, pre-BSx, we observed no significant differences in fecal MPs between groups. However, fecal MP fibers and total MPs positively correlated with portal and total macrophages and total killer T cells while total fecal MPs were positively correlated with natural killer cells. Additionally, 19 genes related to immune system and apoptosis correlated with fecal MPs at baseline. Fecal MP fibers correlated positively with fecal Bifidobacterium and negatively with Lachnospiraceae. Patients with MASH (n = 11) were re-assessed 12-months post-bariatric surgery (BSx) and we found that those with persistent disease (n = 4) had higher fecal MP fragments than those with normalized liver histology (n = 7). At 12-month post-BSx, MP fragments positively correlated with helper T cells and total MPs positively correlated with natural killer T cells and B cells. Our study is the first to look at 1) the role of MPs in MASH and its association with IM, immune cells and hepatic gene expression and 2) look at the role of MPs longitudinally in MASH persistence following BSx. Future research should further explore this relationship.
Collapse
Affiliation(s)
| | - Yasaman Ghorbani
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | | | - Lina Chen
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Kristina M Chomiak
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Anna Christina Tyler
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Nathan C Eddingsaas
- School of Chemistry and Materials Science, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Sandra E Fischer
- Toronto General Hospital, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Timothy D Jackson
- Division of Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Allan Okrainec
- Division of Surgery, University of Toronto, Toronto, Canada; Division of General Surgery, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Medicine, Division of Gastroenterology, University of Toronto, Toronto, Canada.
| |
Collapse
|
44
|
Ruathong W, Khuituan P, Peerakietkhajorn S, Teanpaisan R, Nopparat J. The probiotic Lacticaseibacillus rhamnosus SD11 alleviates the progression of liver and colon damage through modulation of inflammation and tight junction proteins in streptozotocin-induced diabetic mice. PLoS One 2024; 19:e0313395. [PMID: 39570868 PMCID: PMC11581286 DOI: 10.1371/journal.pone.0313395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Lacticaseibacillus rhamnosus SD11 (SD11) has several health benefits for the host, including antidiabetic, anti-inflammatory, and antimicrobial effects. However, the antidiabetic mechanism of SD11 has not been clearly elucidated. The current study assessed the effects of SD11 and the associated underlying mechanisms on streptozotocin (STZ)-induced diabetic mice. Compared with the normal control, SD11 supplementation for 4 weeks significantly improved the metabolic profiles, including body weight (BW), fasting blood glucose (FBG), fasting insulin level (FIN), and liver index (LI), in conjunction with a lower NAS score. A notable reduction in the liver function parameters aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP) and total cholesterol (TC), together with histopathology studies, supported diabetic recovery by SD11. A closer examination of two major markers for the insulin pathway, insulin receptor (INSR) and insulin substrate (IRS)-1, revealed that SD11 could exert its glucose control through the upregulation of these molecules, which were almost demolished in nontreated diabetic livers. Additionally, SD11-treated mice exhibited alleviation of oxidative stress enzymes; downregulation of proinflammatory cytokines, including interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ; and decreased infiltration of macrophages into liver tissue. These findings were concomitant with the preservation of the tight junction proteins occludin and zona occludin (ZO)-1, which in turn lowered the levels of the inflammatory cytokines IL-1β and TNF-α and prevented colon tissue injury to some extent. Notably, the results for the SD11 control mice were identical to those for the normal control mice. Overall, our findings that SD11 delays liver deterioration and reduces colon lesions in diabetic mice provide evidence for the use of SD11 as an effective strategy to improve diabetes-related symptoms.
Collapse
Affiliation(s)
- Waraporn Ruathong
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Pissared Khuituan
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
| | - Saranya Peerakietkhajorn
- Faculty of Science, Division of Biological Science, Prince of Songkla University, Songkhla, Thailand
| | - Rawee Teanpaisan
- Faculty of Dentistry, Research Center of Excellence for Oral Health, Prince of Songkla University, Hat Yai, Thailand
| | - Jongdee Nopparat
- Faculty of Science, Division of Health and Applied Sciences, Prince of Songkla University, Songkhla, Thailand
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
45
|
Grabeklis SA, Kozlova MA, Mikhaleva LM, Dygai AM, Vandysheva RA, Anurkina AI, Areshidze DA. Effect of Constant Illumination on the Morphofunctional State and Rhythmostasis of Rat Livers at Experimental Toxic Injury. Int J Mol Sci 2024; 25:12476. [PMID: 39596541 PMCID: PMC11594381 DOI: 10.3390/ijms252212476] [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: 10/17/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
The effect of dark deprivation on the morphofunctional state and rhythmostasis of the liver under CCl4 toxic exposure has been studied. The relevance of this study is due to the fact that the hepatotoxic effect of carbon tetrachloride on the liver is well studied, but there are very few data on the relationship between CCl4 intoxication and circadian biorhythms, and most of the studies consider the susceptibility of the organism in general and of the liver in particular to the influence of CCl4 in some separate periods of the rhythm, but not the influence of this chemical agent on the structure of the whole rhythm. In addition, earlier studies indicate that light disturbance causes certain changes in the morphofunctional state of the liver and the structure of the circadian rhythm of a number of parameters. As a result of this study, we found that the effect of CCl4 in conditions of prolonged dark deprivation causes more significant structural and functional changes in hepatocytes, as well as leading to significant changes in the circadian rhythms of a number of parameters, which was not observed in the action of CCl4 as a monofactor. We assume that the severity of structural and functional changes is due to the light-induced deficiency of melatonin, which has hepatoprotective properties. Thus, the mechanisms of CCl4 action on CRs under conditions of light regime violations leave a large number of questions requiring further study, including the role of melatonin in these processes.
Collapse
Affiliation(s)
- Sevil A. Grabeklis
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia
| | - Maria A. Kozlova
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia
| | - Lyudmila M. Mikhaleva
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia
| | - Alexander M. Dygai
- Research Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Rositsa A. Vandysheva
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia
| | - Anna I. Anurkina
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia
| | - David A. Areshidze
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, 117418 Moscow, Russia
| |
Collapse
|
46
|
Fujii I, Matsumoto N, Ogawa M, Konishi A, Kaneko M, Watanabe Y, Masuzaki R, Kogure H, Koizumi N, Sugitani M. Artificial Intelligence and Image Analysis-Assisted Diagnosis for Fibrosis Stage of Metabolic Dysfunction-Associated Steatotic Liver Disease Using Ultrasonography: A Pilot Study. Diagnostics (Basel) 2024; 14:2585. [PMID: 39594250 PMCID: PMC11593288 DOI: 10.3390/diagnostics14222585] [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: 09/29/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Elastography increased the diagnostic accuracy of liver fibrosis. However, several challenges persist, including the widespread utilization of equipment, difficulties in measuring certain cases, and the influence of viscosity factors. A rough surface and a blunted hepatic margin have long been acknowledged as valuable characteristics indicative of hepatic fibrosis. The objective of this study was to conduct an image analysis and quantitative assessment of the contour of the sagittal section of the left lobe of the liver. METHODS Between February and October 2020, 486 consecutive outpatients underwent ultrasound examinations at our hospital. A total of 214 images were manually annotated by delineating the liver contour to create annotation images. U-Net was employed for liver segmentation, with the dataset divided into training (n = 128), testing (n = 42), and validation (n = 44) subsets. Additionally, 43 Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) cases with pathology data from between 2015 and 2020 were included. Segmentation was performed using the program developed in the first step. Subsequently, shape analysis was conducted using ImageJ. RESULTS Liver segmentation exhibited high accuracy, as indicated by Dice loss of 0.044, Intersection over Union of 0.935, and an F score of 0.966. The accuracy of the classification of the liver surface as smooth or rough via ResNet 50 was 84.6%. Image analysis showed MinFeret and Minor correlated with liver fibrosis stage (p = 0.046, 0.036, respectively). Sensitivity, specificity, and AUROC of Minor for ≥F3 were 0.571, 0.862, and 0.722, respectively, and F4 were 1, 0.600, and 0.825, respectively. CONCLUSION Deep learning segmentation of the sagittal cross-sectional contour of the left lobe of the liver demonstrated commendable accuracy. The roughness of the liver surface was correctly judged by artificial intelligence. Image analysis showed the thickness of the left lobe inversely correlated with liver fibrosis stage.
Collapse
Affiliation(s)
- Itsuki Fujii
- Department of Mechanical Engineering and Intelligent Systems, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu 182-8585, Japan
| | - Naoki Matsumoto
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Masahiro Ogawa
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Aya Konishi
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Masahiro Kaneko
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Yukinobu Watanabe
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Ryota Masuzaki
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Hirofumi Kogure
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan; (M.O.); (Y.W.)
| | - Norihiro Koizumi
- Department of Mechanical Engineering and Intelligent Systems, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu 182-8585, Japan
| | - Masahiko Sugitani
- Division of Pathology, Nihon University School of Medicine, Tokyo 173-8610, Japan;
| |
Collapse
|
47
|
Hudson D, Afzaal T, Bualbanat H, AlRamdan R, Howarth N, Parthasarathy P, AlDarwish A, Stephenson E, Almahanna Y, Hussain M, Diaz LA, Arab JP. Modernizing metabolic dysfunction-associated steatotic liver disease diagnostics: the progressive shift from liver biopsy to noninvasive techniques. Therap Adv Gastroenterol 2024; 17:17562848241276334. [PMID: 39553445 PMCID: PMC11565685 DOI: 10.1177/17562848241276334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 07/27/2024] [Indexed: 11/19/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health concern worldwide. Liver biopsy is the gold standard for diagnosing and staging MASLD, but it is invasive and carries associated risks. In recent years, there has been significant progress in developing noninvasive techniques for evaluation. This review article discusses briefly current available noninvasive assessments and the various liver biopsy techniques available for MASLD, including invasive techniques such as transjugular and transcutaneous needle biopsy, intraoperative/laparoscopic biopsy, and the evolving role of endoscopic ultrasound-guided biopsy. In addition to discussing the various biopsy techniques, we review the current state of knowledge on the histopathologic evaluation of MASLD, including the various scoring systems used to grade and stage the disease. We also explore current and alternative modalities for histopathologic evaluation, such as whole slide imaging and the utility of immunohistochemistry. Overall, this review article provides a comprehensive overview of the progress in liver biopsy techniques for MASLD and compares invasive and noninvasive modalities. However, beyond clinical trials, the practical application of liver biopsy may be limited, as ongoing advancements in noninvasive fibrosis assessments are expected to more effectively identify candidates for MASLD treatment in real-world settings.
Collapse
Affiliation(s)
- David Hudson
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Tamoor Afzaal
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Hasan Bualbanat
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Raaed AlRamdan
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Nisha Howarth
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Pavithra Parthasarathy
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Alia AlDarwish
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Emily Stephenson
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Yousef Almahanna
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Maytham Hussain
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Luis Antonio Diaz
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- MASLD Research Center, Division of MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
| | - Juan Pablo Arab
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, 1201 E. Broad St. P.O. Box 980341, Richmond, VA 23284, USA
| |
Collapse
|
48
|
Martínez-Montoro JI, Arranz-Salas I, Gutiérrez-Repiso C, Sánchez-García A, Ocaña-Wilhelmi L, Pinazo-Bandera JM, Fernández-García D, Muñoz-Garach A, Morales-García D, García-Cortés M, García-Fuentes E, Tinahones FJ, Garrido-Sánchez L. Weight Loss After Sleeve Gastrectomy According to Metabolic Dysfunction-Associated Steatotic Liver Disease Stage in Patients with Obesity: A Liver Biopsy-Based Prospective Study. Nutrients 2024; 16:3857. [PMID: 39599643 PMCID: PMC11597773 DOI: 10.3390/nu16223857] [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: 10/04/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The role of metabolic dysfunction-associated steatotic liver disease (MASLD) in sleeve gastrectomy (SG)-related outcomes remains uncertain. In this study, we aimed to assess the influence of preoperative biopsy-proven MASLD and its stages on weight loss after SG. METHODS One hundred sixty-three patients with obesity undergoing SG with concomitant intraoperative liver biopsy were followed up for 1 year. Fifty-eight participants were categorized as no MASLD, thirty-eight as metabolic dysfunction-associated steatotic liver (MASL), and sixty-seven as metabolic dysfunction-associated steatohepatitis (MASH). Percentage total weight loss (%TWL) and percentage excess weight loss (%EWL) 1 year after SG were calculated for the different groups. We also evaluated the association between preoperative MASLD (and its stages) and weight loss, after adjusting for potential confounders. RESULTS Significant differences among groups were detected in %EWL (p = 0.004, ANOVA test), but not in %TWL (p = 0.079). However, significant differences in %TWL were found when MASH and no MASH (i.e., participants with MASL and participants without MASLD) groups were compared (27.3 ± 9.9 vs. 30.7 ± 9, respectively, p = 0.025). In the linear regression model for predicting %EWL 1 year after SG, the presence of MASH was independently associated with a lower %EWL, after adjusting for age, sex, baseline body mass index (BMI), and baseline glycated hemoglobin (HbA1c) (Beta -7.1; 95% CI -13.6, -0.5; p = 0.035). The presence of MASLD, liver fibrosis, or advanced liver fibrosis (≥F2) was also associated with lower %EWL after SG in crude models, although they did not remain significant after adjusting for these confounders. The presence of MASH was inversely related to %TWL, although the association did not remain significant after adjustment (Beta -2.7; 95% CI -5.7, 0.2; p = 0.069). CONCLUSIONS MASH may be independently associated with lower %EWL 1 year after SG in patients with obesity.
Collapse
Affiliation(s)
- José Ignacio Martínez-Montoro
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Isabel Arranz-Salas
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Department of Anatomical Pathology, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Department of Human Physiology, Human Histology, Anatomical Pathology and Physical Education, University of Málaga, 29010 Málaga, Spain
| | - Carolina Gutiérrez-Repiso
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana Sánchez-García
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
| | - Luis Ocaña-Wilhelmi
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Department of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
| | - José M. Pinazo-Bandera
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Department of Gastroenterology, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Diego Fernández-García
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Araceli Muñoz-Garach
- Department of Endocrinology and Nutrition, Virgen de las Nieves University Hospital, 18014 Granada, Spain
| | - Dieter Morales-García
- Department of General and Digestive Surgery, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
| | - Miren García-Cortés
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Department of Gastroenterology, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Eduardo García-Fuentes
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Department of Gastroenterology, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine and Dermatology, Faculty of Medicine, University of Málaga, 29010 Málaga, Spain
| | - Lourdes Garrido-Sánchez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
49
|
Soluyanova P, Quintás G, Pérez-Rubio Á, Rienda I, Moro E, van Herwijnen M, Verheijen M, Caiment F, Pérez-Rojas J, Trullenque-Juan R, Pareja E, Jover R. The Development of a Non-Invasive Screening Method Based on Serum microRNAs to Quantify the Percentage of Liver Steatosis. Biomolecules 2024; 14:1423. [PMID: 39595599 PMCID: PMC11592063 DOI: 10.3390/biom14111423] [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: 10/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is often asymptomatic and underdiagnosed; consequently, there is a demand for simple, non-invasive diagnostic tools. In this study, we developed a method to quantify liver steatosis based on miRNAs, present in liver and serum, that correlate with liver fat. The miRNAs were analyzed by miRNAseq in liver samples from two cohorts of patients with a precise quantification of liver steatosis. Common miRNAs showing correlation with liver steatosis were validated by RT-qPCR in paired liver and serum samples. Multivariate models were built using partial least squares (PLS) regression to predict the percentage of liver steatosis from serum miRNA levels. Leave-one-out cross validation and external validation were used for model selection and to estimate predictive performance. The miRNAseq results disclosed (a) 144 miRNAs correlating with triglycerides in a set of liver biobank samples (n = 20); and (b) 124 and 102 miRNAs correlating with steatosis by biopsy digital image and MRI analyses, respectively, in liver samples from morbidly obese patients (n = 24). However, only 35 miRNAs were common in both sets of samples. RT-qPCR allowed to validate the correlation of 10 miRNAs in paired liver and serum samples. The development of PLS models to quantitatively predict steatosis demonstrated that the combination of serum miR-145-3p, 122-5p, 143-3p, 500a-5p, and 182-5p provided the lowest root mean square error of cross validation (RMSECV = 1.1, p-value = 0.005). External validation of this model with a cohort of mixed MASLD patients (n = 25) showed a root mean squared error of prediction (RMSEP) of 5.3. In conclusion, it is possible to predict the percentage of hepatic steatosis with a low error rate by quantifying the serum level of five miRNAs using a cost-effective and easy-to-implement RT-qPCR method.
Collapse
Affiliation(s)
- Polina Soluyanova
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Guillermo Quintás
- Health and Biomedicine, LEITAT Technological Center, 08225 Terrassa, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, 28029 Madrid, Spain
| | - Álvaro Pérez-Rubio
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Iván Rienda
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (I.R.); (J.P.-R.)
| | - Erika Moro
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Marcel van Herwijnen
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Marcha Verheijen
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Florian Caiment
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Judith Pérez-Rojas
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (I.R.); (J.P.-R.)
| | - Ramón Trullenque-Juan
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Eugenia Pareja
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Ramiro Jover
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
50
|
Farías C, Cisternas C, Caicedo A, Mercado L, Valenzuela R, Calderón H, Espinosa A, Videla LA, Muñoz LA. High-fiber basil seed flour reduces insulin resistance and hepatic steatosis in high-fat diet mice. NPJ Sci Food 2024; 8:90. [PMID: 39516211 PMCID: PMC11549410 DOI: 10.1038/s41538-024-00329-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
The incidence of insulin resistance (IR) and hepatic steatosis is increasing, with dietary fiber playing a protective role against these disorders. Ocimum basilicum L., widely used in food, pharmaceutical, and cosmetic industries, but their health-promoting properties remain underexplored. This study evaluated the effects of a fiber-rich fraction of partially defatted basil seeds (BSF) on IR, hepatic steatosis, and polyunsaturated fatty acid and short-chain fatty acid (SCFA) profiles in high-fat diet (HFD)-fed C57BL/6 J male mice. Mice were assigned to four groups and fed either a control diet or HFD, supplemented with BSF or oat flour for 4 weeks. HFD induced IR, hepatic steatosis, proinflammatory state, and a significant decreased in SCFA production. In contrast, supplementation with BSF attenuated IR, steatosis, liver damage, oxidative stress, and inflammation, while increasing n-3 polyunsaturated fatty acids in liver, adipocytes, and erythrocytes, and enhancing SCFA production, suggesting potential therapeutic benefits in managing these conditions.
Collapse
Affiliation(s)
- Camila Farías
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, 8380000, Chile
| | - Camila Cisternas
- School of Health Care Sciences , Universidad San Sebastián, Puerto Montt, Chile
| | - Angie Caicedo
- School of Agronomy, Faculty of Agronomy and Food Sciences , Pontificia Universidad Católica de Valparaíso, Quillota, 2260000, Chile
| | - Lorena Mercado
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, 8380000, Chile
- Universidad Andrés Bello, Medicina, Facultad Medicina, 8370035, Santiago, Chile
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, 8380000, Chile
| | - Héctor Calderón
- Food Science Lab, Faculty of Medicine and Health Sciences, Universidad Central de Chile, Santiago, 8330546, Chile
| | - Alejandra Espinosa
- Center of Interdisciplinary Biomedical and Engineering Research for Health-MEDING. Universidad de Valparaíso, Valparaíso, Chile
- Medical Technology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - L A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Loreto A Muñoz
- Food Science Lab, Faculty of Medicine and Health Sciences, Universidad Central de Chile, Santiago, 8330546, Chile.
| |
Collapse
|