1
|
Xiao L, Zeng L, Wang J, Hong C, Zhang Z, Wu C, Cui H, Li Y, Li R, Liang S, Deng Q, Li W, Zou X, Ma P, Liu L. Development and Validation of Machine Learning-Based Marker for Early Detection and Prognosis Stratification of Nonalcoholic Fatty Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e10527. [PMID: 40432473 DOI: 10.1002/advs.202410527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 04/27/2025] [Indexed: 05/29/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease and is considered the hepatic manifestation of metabolic syndrome, triggering out adverse outcomes. A stacked multimodal machine learning model is constructed and validated for early identification and prognosis stratification of NAFLD by integrating genetic and clinical data sourced from 36 490 UK Biobank and 9 007 Nanfang Hospital participants and extracted its probabilities as in-silico scores for NAFLD (ISNLD). The efficacy of ISNLD is evaluated for the early prediction of severe liver disease (SeLD) and analyzed its association with metabolism-related outcomes. The multimodal model performs satisfactorily in classifying individuals into low- and high-risk groups for NAFLD, achieving area under curves (AUCs) of 0.843, 0.840, and 0.872 within training, internal, and external test sets, respectively. Among high-risk group, ISNLD is significantly associated with intrahepatic and metabolism-related complications after lifestyle factors adjustment. Further, ISNLD demonstrates notable capability for early prediction of SeLD and further stratifies high-risk subjects into three risk subgroups of elevated risk for adverse outcomes. The findings emphasize the model's ability to integrate multimodal features to generate ISNLD, enabling early detection and prognostic prediction of NAFLD. This facilitates personalized stratification for NAFLD and metabolism-related outcomes based on digital non-invasive markers, enabling preventive interventions.
Collapse
Affiliation(s)
- Lushan Xiao
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lin Zeng
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518133, China
| | - Jiaren Wang
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chang Hong
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziyong Zhang
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengkai Wu
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Cui
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ruining Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shengxing Liang
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qijie Deng
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenyuan Li
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xuejing Zou
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pengcheng Ma
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Liu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
2
|
Cho JH, Kim HG, Huang M, Wang S, Liu S, Lu A, McCrocklin K, Zhang Y, Fang Z, Wang J, Liu W, Wan J, Dong XC. The Patatin-Like Phospholipase Domain-Containing 3 148M Variant Exacerbates Alcohol-Induced Liver Injury and Tumorigenesis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00154-3. [PMID: 40350061 DOI: 10.1016/j.ajpath.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/02/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Patatin-like phospholipase domain-containing 3 (PNPLA3) protein 148M variant is strongly associated with cirrhosis and hepatocellular carcinoma (HCC); however, the underlying mechanisms remain elusive. This study aimed to elucidate the role of the PNPLA3148M variant in alcohol-related HCC development. Control and humanized PNPLA3148M transgenic mice were fed with an ethanol-containing diet for 12 weeks. The animals were examined for liver tumors. After the alcohol feeding, the PNPLA3148M mice had twofold higher liver cancer incidence rates and larger tumor sizes than those in the control mice. Cancer stem cell markers in the PNPLA3148M mouse livers were elevated relative to those in the control mouse livers. Alcohol detoxification was impaired in the PNPLA3148M mouse livers. Hepatic oxidative stress and DNA damage were elevated in the PNPLA3148M mice. Wnt/β-catenin and Yes-associated protein (YAP) and WW domain-containing transcription regulator 1 (TAZ) were activated in the PNPLA3148M mouse livers. The data suggest that the PNPLA3148M variant has a strong interaction with alcohol in HCC development through attenuation of alcohol detoxification and promotion of oncogenic pathways. Targeting the PNPLA3148M variant might be useful for the prevention or treatment of alcohol-associated HCC in patients carrying this variant.
Collapse
Affiliation(s)
- Jung-Hyo Cho
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana; Department of East and West Cancer Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Hyeong-Geug Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana; Center for Diabetes and Metabolic Diseases, Indiana University, Indianapolis, Indiana; Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana
| | - Shen Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Sheng Liu
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Alex Lu
- Park Tudor School, Indianapolis, Indiana
| | - Kyle McCrocklin
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana
| | - Yang Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Zhigang Fang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Juexin Wang
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Jun Wan
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana; Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, Indiana
| | - X Charlie Dong
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana; Center for Diabetes and Metabolic Diseases, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana; Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
3
|
Yun J, Min YS. Association Between Perfluoroalkyl Substance (PFAS) Exposure and Nonalcoholic Fatty Liver Disease in Korean Adults: Results From the KoNEHS 2018-2020: A Cross-Sectional Study. Am J Ind Med 2025. [PMID: 40341549 DOI: 10.1002/ajim.23732] [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: 09/19/2024] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025]
Abstract
OBJECTIVES Nonalcoholic fatty liver disease (NAFLD) is a major public health problem and the most common chronic liver disease today. In Korea, the prevalence and incidence of NAFLD are currently very high, causing a serious social burden. Perfluoroalkyl substances (PFAS) have been consistently implicated as a potential cause of NAFLD, but research in Koreans is limited. METHODS Using data from the 4th Korean National Environmental Health Survey (KoNEHS, n = 2859), we investigated the association between PFAS blood levels and NAFLD. Multivariable logistic regression models were used to calculate odds ratios (ORs) for the effects of PFAS. A mediation analysis was also conducted to examine the mediating effect of obesity. Finally, weighted quantile sum (WQS) and G-computation methods were implemented to evaluate the joint effect of PFAS mixtures. Hepatic steatosis index was used as a diagnostic tool for NAFLD. RESULTS Through multivariable logistic regression, statistically significant associations with NAFLD were observed for perfluorooctanoic acid (PFOA) (OR 1.09-1.39), perfluorooctansulfonate (PFOS) (1.09-1.40), perfluorohexanesulfonic acid (PFHxS) (1.04-1.22), perfluorononanoic acid (PFNA) (1.12-1.42), and total PFAS (1.21-1.81). We also found that obesity was a significant mediator for PFOA, PFNA, and total PFAS. The ORs for NAFLD obtained by WQS and G-computation methods in the multivariable adjusted model were 1.10-1.46 and 1.08-1.32, respectively. CONCLUSIONS This study confirmed a significant association between some PFAS and increased odds of NAFLD. Excessive exposure to PFAS might explain the high prevalence and incidence of chronic liver disease in Koreans. Long-term cohort studies are needed to assess geographic and occupational exposures in the Korean population.
Collapse
Affiliation(s)
- Jisuk Yun
- Department of Occupational and Environmental Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
- Department of Statistics and Data Science, Korea National Open University, Korea
| | - Young-Sun Min
- Department of Occupational and Environmental Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| |
Collapse
|
4
|
Cao R, Zhang Y, Cao L, Jiang H. Is type 2 diabetes a link between lung function and metabolic dysfunction-associated steatotic liver disease? Insights from population studies and Mendelian randomization. Eur J Gastroenterol Hepatol 2025; 37:652-659. [PMID: 39976012 DOI: 10.1097/meg.0000000000002941] [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] [Indexed: 02/21/2025]
Abstract
AIM This study aimed to investigate the relationship between lung function and metabolic dysfunction-associated steatotic liver disease (MASLD), and the potential mediating role of type 2 diabetes. METHODS Data from the 2007 to 2012 National Health and Nutrition Examination Survey were used. Logistic regression analysis was employed to assess the association between lung function parameters [forced vital capacity (FVC), forced expiratory volume in 1 s (FEV 1 ), FEV 1 /FVC] and MASLD prevalence while exploring type 2 diabetes mediation. Further analyses included linkage disequilibrium score regression, Mendelian randomization, and meta-analysis to examine the causal relationship between lung function and MASLD, considering type 2 diabetes mediation. RESULTS The results showed that higher FVC and FEV 1 levels were associated with decreased MASLD risk, with type 2 diabetes partially mediating this relationship. Genetic analyses supported a causal link between lung function and MASLD, with type 2 diabetes acting as an intermediary. However, no significant association was found between FEV 1 /FVC and MASLD. CONCLUSION The study identified a causal relationship between lung function and MASLD, with type 2 diabetes playing a partial mediating role.
Collapse
Affiliation(s)
- Runmin Cao
- Graduate School, Jinzhou Medical University (Jinzhou Central Hospital), Jinzhou, Liaoning Province
| | - Yurun Zhang
- Department of Rehabilitation Therapy, Shandong Xiandai University, Jinan, Shandong Province
| | - Ling Cao
- Department of Chronic Disease Prevention and Control, Jieshou City Center for Disease Control and Prevention, Fuyang
| | - Honghe Jiang
- Department of Clinical Medicine, Anhui University of Science and Technology, Huainan, Anhui Province, China
| |
Collapse
|
5
|
Huang DQ, Wong VWS, Rinella ME, Boursier J, Lazarus JV, Yki-Järvinen H, Loomba R. Metabolic dysfunction-associated steatotic liver disease in adults. Nat Rev Dis Primers 2025; 11:14. [PMID: 40050362 DOI: 10.1038/s41572-025-00599-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 03/09/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the umbrella term that comprises metabolic dysfunction-associated steatotic liver, or isolated hepatic steatosis, through to metabolic dysfunction-associated steatohepatitis, the progressive necroinflammatory disease form that can progress to fibrosis, cirrhosis and hepatocellular carcinoma. MASLD is estimated to affect more than one-third of adults worldwide. MASLD is closely associated with insulin resistance, obesity, gut microbial dysbiosis and genetic risk factors. The obesity epidemic and the growing prevalence of type 2 diabetes mellitus greatly contribute to the increasing burden of MASLD. The treatment and prevention of major metabolic comorbidities such as type 2 diabetes mellitus and obesity will probably slow the growth of MASLD. In 2023, the field decided on a new nomenclature and agreed on a set of research and action priorities, and in 2024, the US FDA approved the first drug, resmetirom, for the treatment of non-cirrhotic metabolic dysfunction-associated steatohepatitis with moderate to advanced fibrosis. Reliable, validated biomarkers that can replace histology for patient selection and primary end points in MASH trials will greatly accelerate the drug development process. Additionally, noninvasive tests that can reliably determine treatment response or predict response to therapy are warranted. Sustained efforts are required to combat the burden of MASLD by tackling metabolic risk factors, improving risk stratification and linkage to care, and increasing access to therapeutic agents and non-pharmaceutical interventions.
Collapse
Affiliation(s)
- Daniel Q Huang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore
| | - Vincent W S Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Mary E Rinella
- University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Jerome Boursier
- Service d'Hépato-Gastroentérologie et Oncologie Digestive, Centre Hospitalier Universitaire d'Angers, Angers, France
- Laboratoire HIFIH, SFR ICAT 4208, Université d'Angers, Angers, France
| | - Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, San Diego, CA, USA.
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California at San Diego, San Diego, CA, USA.
| |
Collapse
|
6
|
Zhang J, Yang W, Zhu Y, Li Z, Zheng Y, Zhang Y, Gao W, Zhang X, Wu Z, Gao L. Microenvironment-induced programmable nanotherapeutics restore mitochondrial dysfunction for the amelioration of non-alcoholic fatty liver disease. Acta Biomater 2025; 194:323-335. [PMID: 39805524 DOI: 10.1016/j.actbio.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a metabolic liver disorder with severe complications. Mitochondrial dysfunction due to over-opening of the mitochondrial permeability transition pore (mPTP) in liver cells plays a central role in the development and progression of NAFLD. Restoring mitochondrial function is a promising strategy for NAFLD therapy. Herein, we designed and developed a microenvironment-induced programmable nanotherapeutic to restore mitochondrial function and ameliorate NAFLD. cyclosporine (Cyclosporine capsules) A (CsA), as a highly effective inhibitors of the opening of mPTP, was chosen in the present work. Nanotherapeutics were prepared by assembling two structurally simple multifunctional glucosamine derivatives: dextran-grafted galactose (Dex-Gal) and Dex-triphenylphosphine (Dex-TPP). Galactose units in the nanotherapeutics guide the hepatocyte-specific uptake. Detachment of galactose from acidic lysosomes via Schiff base cleavage exposes the TPP moieties, which subsequently steers the nanotherapeutics to escape from lysosomes and target mitochondria through an enhanced positive charge, enabling precise in situ drug delivery. Simultaneously, the nanotherapeutics improved mitochondrial dysfunction by inhibiting palmitic acid-induced opening of the mitochondrial permeability transition pore in HepG2 cells, maintaining mitochondrial membrane potential, and decreasing reactive oxygen species production. Furthermore, CsA@Dex-Gal/TPP accumulated in the livers of NAFLD mice, restored mitochondrial autophagy, regulated abnormalities in glucose and lipid metabolism, and improved hepatic lipid deposition. This study offers a new cascading strategy for targeting liver cell mitochondria to treat NAFLD and other mitochondria-associated diseases. STATEMENT OF SIGNIFICANCE: We design microenvironment-induced programmable nanotherapeutics for NAFLD Nanotherapeutics has the capabilities of lysosomal escape and mitochondrial targeting Nanotherapeutics improves mitochondrial dysfunction and ameliorates NAFLD This study offers a new cascading strategy for other mitochondria-associated diseases.
Collapse
Affiliation(s)
- Jun Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Wenyi Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Yue Zhu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Zhanbin Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China
| | - Yin Zheng
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Weisong Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhongming Wu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
7
|
Grune E, Nattenmüller J, Kiefer LS, Machann J, Peters A, Bamberg F, Schlett CL, Rospleszcz S. Subphenotypes of body composition and their association with cardiometabolic risk - Magnetic resonance imaging in a population-based sample. Metabolism 2025; 164:156130. [PMID: 39743039 DOI: 10.1016/j.metabol.2024.156130] [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: 10/01/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND For characterizing health states, fat distribution is more informative than overall body size. We used population-based whole-body magnetic resonance imaging (MRI) to identify distinct body composition subphenotypes and characterize associations with cardiovascular disease (CVD) risk. METHODS Bone marrow, visceral, subcutaneous, cardiac, renal, hepatic, skeletal muscle and pancreatic adipose tissue were measured by MRI in n = 299 individuals from the population-based KORA cohort. Body composition subphenotypes were identified by data-driven k-means clustering. CVD risk was calculated by established scores. RESULTS We identified five body composition subphenotypes, which differed substantially in CVD risk factor distribution and CVD risk. Compared to reference subphenotype I with favorable risk profile, two high-risk phenotypes, III&V, had a 3.8-fold increased CVD risk. High-risk subphenotype III had increased bone marrow and skeletal muscle fat (26.3 % vs 11.4 % in subphenotype I), indicating ageing effects, whereas subphenotype V showed overall high fat contents, and particularly elevated pancreatic fat (25.0 % vs 3.7 % in subphenotype I), indicating metabolic impairment. Subphenotype II had a 2.7-fold increased CVD risk, and an unfavorable fat distribution, probably smoking-related, while BMI was only slightly elevated. Subphenotype IV had a 2.8-fold increased CVD risk with comparably young individuals, who showed high blood pressure and hepatic fat (17.7 % vs 3.0 % in subphenotype I). CONCLUSIONS Whole-body MRI can identify distinct body composition subphenotypes associated with different degrees of cardiometabolic risk. Body composition profiling may enable a more comprehensive risk assessment than individual fat compartments, with potential benefits for individualized prevention.
Collapse
Affiliation(s)
- Elena Grune
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany; Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Johanna Nattenmüller
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Radiology and Nuclear Medicine Hirslanden Clinic St. Anna, Lucerne, Switzerland
| | - Lena S Kiefer
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, Eberhard Karls University of Tuebingen, Tuebingen, Germany; Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Jürgen Machann
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany; Institute for Diabetes Research and Metabolic Diseases, Helmholtz Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Medical Faculty, Ludwig-Maximilians-Universität (LMU), Munich, Germany; German Center for Cardiovascular Disease Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christopher L Schlett
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susanne Rospleszcz
- Department of Diagnostic and Interventional Radiology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Epidemiology, Helmholtz Munich, Neuherberg, Germany.
| |
Collapse
|
8
|
Liu Z, Chen X, Yuan H, Jin L, Zhang T, Chen X. Dissecting the shared genetic architecture between nonalcoholic fatty liver disease and type 2 diabetes. Hum Mol Genet 2025; 34:338-346. [PMID: 39690818 DOI: 10.1093/hmg/ddae184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Observational studies have reported a bidirectional correlation between nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), but the shared genetic basis between the two conditions remains unclear. Using genome-wide association study (GWAS) summary data from European-ancestry populations, we examined the cross-trait genetic correlation and identified genomic overlaps and shared risk loci. We employed a latent causal variable model and Mendelian randomization (MR) analysis to infer causal relationships. Colocalization analysis and conditional/conjunctional false discovery rate (condFDR/conjFDR) were used to identify genomic overlaps and shared risk loci. Two-step MR analysis was utilized to identify potential mediators. We observed a strong positive genomic correlation between NAFLD and T2D (rg = 0.652, P = 5.67 × 10-6) and identified tissue-specific transcriptomic correlations in the pancreas, liver, skeletal muscle, subcutaneous adipose, and blood. Genetic enrichment was observed in NAFLD conditional on associations with T2D and vice versa, indicating significant polygenic overlaps. We found robust evidence for the causal effect of NAFLD on T2D, particularly insulin-related T2D, rather than vice versa. Colocalization analysis identified shared genomic regions between NAFLD and T2D, including GCKR, FTO, MAU2-TM6SF2, and PNPLA3-SAMM50. High-density lipoprotein cholesterol and insulin were partly mediated the association between NAFLD and T2D. These findings unveil a close genetic link between NAFLD and T2D, shedding light on the biological mechanisms connecting NAFLD progression to T2D.
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Xiaochen Chen
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South RD, Shanghai 200025, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, 130 Dong'an RD, Shanghai 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, 130 Dong'an RD, Shanghai 200032, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Urumqi RD, Shanghai 200040, China
| |
Collapse
|
9
|
Wei S, Jiang W, Zheng H, Zhang J, Yang J, Wang Y, Liu Y, Sun L, Li X, Wei J, Sun W. The combined impact of BMI and ABSI on all-cause mortality among American adults with diabetes. Diabetol Metab Syndr 2025; 17:48. [PMID: 39920852 PMCID: PMC11806875 DOI: 10.1186/s13098-025-01614-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/26/2025] [Indexed: 02/09/2025] Open
Abstract
OBJECTIVE Previous studies have emphasized the independent effects of anthropometric indices-including body mass index (BMI), A Body Shape Index (ABSI), waist-to-height ratio (WHtR), body roundness index (BRI), and Conicity Index-on mortality. However, their combined impact, especially in diabetic populations with distinct obesity patterns, has been less frequently explored. This study investigates both the independent and combined effects of these anthropometric indices on mortality in diabetic Americans and compares their individual and combined diagnostic value. METHODS A nationally representative cohort study was conducted using NHANES data (2005-2018), including 6,572 diabetic adults. Weighted Cox proportional hazards models and restricted cubic splines were applied to evaluate the independent and combined associations of anthropometric indices (BMI, ABSI, WHtR, BRI, and Conicity Index) with all-cause mortality. The weighted receiver operating characteristic (ROC) curve was used to assess the diagnostic value of individual anthropometric indices and their combinations in predicting mortality. RESULTS Among all the anthropometric indices, ABSI exhibited the strongest independent association with all-cause mortality, outperforming other measures such as BMI, WHtR, BRI, and Conicity Index. A clear linear relationship was identified, with higher ABSI tertiles consistently linked to an increased risk of mortality. Notably, within each BMI tertile, ABSI effectively differentiated mortality risk, particularly in the highest tertile. Furthermore, ABSI demonstrated the highest predictive performance among individual metrics (weighted AUC = 0.653) and showed further improvement when combined with BMI (weighted AUC = 0.669). CONCLUSION BMI and ABSI collectively provide a comprehensive evaluation of mortality risk in diabetic populations, capturing the synergistic effects of general and central obesity. These findings highlight the importance of integrating BMI and ABSI into risk assessments to identify high-risk individuals and guide targeted interventions for reducing mortality.
Collapse
Affiliation(s)
- Shuwu Wei
- Beijing University of Chinese Medicine, Beijing, China
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weimin Jiang
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Huijuan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiale Zhang
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Yang
- Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaoxian Wang
- Henan University of Chinese Medicine, Beijing, China
| | - Yang Liu
- Department of Chinese Medicine, Cangzhou Central Hospital, No.1 Cangzhou, Cangzhou, China
| | - Liqiao Sun
- Department of Chinese Medicine, Cangzhou Central Hospital, No.1 Cangzhou, Cangzhou, China
| | - Xinrong Li
- Department of Chinese Medicine, Cangzhou Central Hospital, No.1 Cangzhou, Cangzhou, China
| | - Junping Wei
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weiwei Sun
- Beijing University of Chinese Medicine, Beijing, China.
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
10
|
He Y, Ye M, Xia Y, Zhong Z, Li Q. Antioxidants and the risk of metabolic dysfunction-associated steatotic liver disease: results of National Health and Nutrition Examination Survey and two-sample Mendelian randomization analyses. Eur J Gastroenterol Hepatol 2025; 37:230-239. [PMID: 39621882 DOI: 10.1097/meg.0000000000002898] [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] [Indexed: 12/22/2024]
Abstract
BACKGROUND The link between antioxidants and metabolic dysfunction-associated steatotic liver disease (MASLD) is a topic of considerable discussion in the field of observational studies, with the exact causal connections still being unclear. METHODS In this investigation, a cohort consisting of 17 061 participants from the National Health and Nutrition Examination Surveys was studied. Initially, a cross-sectional analysis was carried out to examine the relationship between the CDAI and MASLD. Further, Mendelian randomization (MR) was utilized to assess the possible causal links between antioxidant levels in the bloodstream and MASLD. RESULTS The association between the CDAI and MASLD was found to be significant in the fully adjusted logistic regression model, showing an OR of 0.95 [95% confidence interval (CI): 0.94-0.97; P < 0.001]. The use of restricted cubic spline regression revealed no significant nonlinear association between the CDAI and the occurrence of MASLD ( Pnonlinearity = 0.321). Additionally, MR findings did not suggest any causal connections between circulating levels of various antioxidants and MASLD. These antioxidants included vitamin A (retinol) (IVW: OR: 0.67, 95% CI: 0.33-1.36, P = 0.272), vitamin C (ascorbate) (IVW: OR: 0.61, 95% CI: 0.34-1.09, P = 0.094), vitamin E (α-tocopherol) (IVW: OR: 0.55, 95% CI: 0.13-2.25, P = 0.407), vitamin E (γ-tocopherol) (IVW: OR: 0.89, 95% CI: 0.36-2.23, P = 0.806), zinc (IVW: OR: 0.95, 95% CI: 0.82-1.09, P = 0.449), selenium (IVW: OR: 0.98, 95% CI: 0.84-1.16, P = 0.855), and carotene (IVW: OR: 0.80, 95% CI: 0.36-1.81, P = 0.596). CONCLUSION The findings highlight a significant negative linear relationship between CDAI and MASLD prevalence in the observational component of the study. However, the MR analysis did not indicate any causal effects of circulating antioxidant levels on MASLD.
Collapse
Affiliation(s)
- Yijia He
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
11
|
Li Y, Ma X, Cui S, Jiang G, Jia J, Ge X, Cao L. Association Between Serum Uric Acid and Non-alcoholic Fatty Liver Disease in Non-obese Young Population: A Prospective Cohort Study. Dig Dis Sci 2025; 70:825-834. [PMID: 39719467 DOI: 10.1007/s10620-024-08808-9] [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: 10/22/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
OBJECTIVE The prevalence of non-alcoholic fatty liver disease (NAFLD) is gradually increasing among non-obese people and shows a trend of younger age. The objective of this study was to investigate the potential association between serum uric acid (SUA) and NAFLD in a non-obese young population. PATIENTS AND METHODS The study recruited 10,938 participants without NAFLD (18 ≤ age < 45,18.5 ≤ BMI < 25) in 2006. After a median follow-up of 9.95 years, 4835 (44.20%) participants were diagnosed with NAFLD. According to the baseline SUA level, the research subjects were divided into 4 groups by the quartile method. The association between SUA and NAFLD was predicted using the Kaplan -Meier survival curve and Cox hazard regression model. RESULTS After adjusting for all the confounders, the HRs and 95%CI for NAFLD in the quartile 2, quartile 3 and quartile 4 were 1.095(1.004 ~ 1.193), 1.195(1.095 ~ 1.304) and 1.409(1.289 ~ 1.541). The restrictive cubic spline (RCS) functions confirmed the existence of a non-linear association between the initial SUA and NAFLD (non-linearity association P < 0.05). Kaplan-Meier Survival Curve shows that the risk of NAFLD increased with increasing levels of SUA. The cumulative incidences of the four groups were 46.19, 51.99, 56.52 and 65.87%, respectively (log-rank test P < 0.001). CONCLUSION It has been confirmed through a prospective cohort study that in non-obese young population, SUA can serve as an independent predictor of NAFLD. An increase in SUA within the physiological range, even if it doesn't reach the level of hyperuricemia, can impose a significant burden on the occurrence of NAFLD. TRIAL REGISTRATION Trial registration number: ChiCTR-TNC-11001489, registration site: http://www.chictr.org.cn/index.aspx .
Collapse
Affiliation(s)
- Yunpeng Li
- Graduate School of North, China University of Science and Technology, Tangshan, China
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Xiangming Ma
- Graduate School of North, China University of Science and Technology, Tangshan, China
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Shuqing Cui
- Graduate School of North, China University of Science and Technology, Tangshan, China
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Guochao Jiang
- Graduate School of North, China University of Science and Technology, Tangshan, China
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Jianguo Jia
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Xinyu Ge
- Graduate School of North, China University of Science and Technology, Tangshan, China
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Liying Cao
- Graduate School of North, China University of Science and Technology, Tangshan, China.
- Department of Hepatobiliary Surgery, Kailuan General Hospital, Tangshan, 063000, China.
| |
Collapse
|
12
|
Perez-Ternero C, Li W, Aubdool AA, Goldin RD, Loy J, Devalia K, Alazawi W, Hobbs AJ. Endogenous C-type natriuretic peptide offsets the pathogenesis of steatohepatitis, hepatic fibrosis, and portal hypertension. PNAS NEXUS 2025; 4:pgae579. [PMID: 39816244 PMCID: PMC11734523 DOI: 10.1093/pnasnexus/pgae579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), hepatic fibrosis, and portal hypertension constitute an increasing public health problem due to the growing prevalence of obesity and diabetes. C-type natriuretic peptide (CNP) is an endogenous regulator of cardiovascular homeostasis, immune cell reactivity, and fibrotic disease. Thus, we investigated a role for CNP in the pathogenesis of MASLD. Wild-type (WT), global CNP (gbCNP-/-), and natriuretic peptide receptor-C (NPR-C-/-) knockout mice were fed a choline-deficient defined amino acid diet or administered CCl4. Liver damage was assessed by histological and biochemical analyses, with steatosis and portal vein size determined by ultrasound. Portal vein pressure and reactivity were measured in vivo and ex vivo, respectively. Pharmacological CNP delivery was used to evaluate prospective therapeutic benefit, and plasma CNP concentration was compared in controls and patients with cirrhosis. Circulating CNP concentration was lower in patients with cirrhosis compared with controls. gbCNP-/- mice were more susceptible, versus WT, to advanced steatohepatitis and hepatic fibrosis, characterized by increased immune cell infiltration, fibrosis, ballooning, plasma alanine aminotransferase concentration, and up-regulation of markers driving these processes. gbCNP-/- mice had increased portal vein diameter and pressure, underpinned by CNP insensitivity. NPR-C-/- animals recapitulated, comparatively, the exaggerated pathogenic phenotype in gbCNP-/- mice, whereas CNP reduced hepatic stellate cell proliferation via NPR-B-dependent inhibition of extracellular signal-related kinase 1/2. Administration of CNP reversed many aspects of disease severity. These data define a new intrinsic role for CNP in offsetting the pathogenesis of MASLD, hepatic fibrosis, and portal hypertension and the potential for targeting CNP signaling for treating these disorders.
Collapse
Affiliation(s)
- Cristina Perez-Ternero
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Aisah A Aubdool
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Robert D Goldin
- Centre for Pathology, St Mary’s Hospital, Imperial College, London W2 1NY, United Kingdom
| | - John Loy
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Adrian J Hobbs
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
13
|
Dinçer BT, Usta AM, Kural A, Helvacı N, Uçar A, Urgancı N. Can fecal calprotectin be used as a biomarker of non-alcoholic fatty liver disease in obese adolescents? BMC Pediatr 2024; 24:834. [PMID: 39716084 PMCID: PMC11665081 DOI: 10.1186/s12887-024-05327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND The incidence of non-alcoholic fatty liver disease (NAFLD) is increasing with obesity, and it is believed that the ongoing low-grade inflammation in obesity and alterations in the enterohepatic axis contributing this process. This study aimed to determine the role of fecal calprotectin (FC) as inflammatory biomarker in obesity and NAFLD. METHODS Between November 2022-August 2023, 31 obese and 10 healthy adolescents aged between 10 and 18 years enrolled in this prospective controlled study. Body mass index higher than 2 standard deviation is considered as obesity. Obese adolescents were divided into two subgroups: obese adolescents (n = 11) and Obese + NAFLD group (n = 20). NAFLD diagnosis was made with biochemical analysis or ultrasonography. FC levels and laboratory parameters analyzed in study group, while only FC samples taken from control group. Anthropometric and laboratory parameters were compared between groups. This study was registered in ClinicalTrials.gov (NCT06229184). RESULTS The median (IQR P25-75) FC levels in the obese + NAFLD, obese and the healthy controls were 136.23 (43.36-332.04), 61.77 (29.70-285.92) and 38.95 (27.59-50.52) µg/g feces, respectively (p = 0.018). Subgroup analyses revealed that the significant difference was between the obese + NAFLD group and the control group (p = 0.02), while no significant differences were observed between the control and obese groups, or between the obese and obese + NAFLD groups. FC positivity rates were 20% (n = 2) in the control group, 54.5% (n = 6) in the obese group, and 75% (n = 15) in the Obese + NAFLD group (p = 0.018). CONCLUSIONS FC is significantly higher in obese adolescents compared to healthy peers, but no significant difference was observed between obese and obese + NAFLD groups. Further studies needed on this subject. TRIAL REGISTRATION This trial is registered in ClinicalTrials.gov (Trial registration number [ClinicalTrials.gov ID] NCT06229184).
Collapse
Affiliation(s)
- Büşra Tetik Dinçer
- Department of Pediatrics, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey.
| | - Ayşe Merve Usta
- Department of Pediatrics, Division of Pediatric Gastroenterology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Alev Kural
- Department of Biochemistry, Bakırkoy Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Nazlı Helvacı
- Department of Biochemistry, Bakırkoy Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ahmet Uçar
- Department of Pediatrics, Division of Pediatric Endocrinology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Nafiye Urgancı
- Department of Pediatrics, Division of Pediatric Gastroenterology, Sisli Hamidiye Etfal Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
14
|
Fan H, Kouvari M, Mingrone G, George J, Papatheodoridis G, Valenzuela-Vallejo L, Liu Z, Chen X, Zhang T, Mantzoros CS. Lipoprotein (a) in the Full Spectrum of Metabolic Dysfunction-associated Steatotic Liver Disease: Evidence From Histologically and Genetically Characterized Cohorts. Clin Gastroenterol Hepatol 2024:S1542-3565(24)01065-6. [PMID: 39672249 DOI: 10.1016/j.cgh.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND & AIMS Lipoprotein(a) (Lp(a)) is an emerging biomarker for cardiometabolic factors. We studied the role of Lp(a) in the full spectrum of metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Three independent analyses were implemented using a multi-center, cross-sectional, liver biopsy-based study (n = 332) (Study 1) and the UK Biobank prospective study (n = 270,004) (Study 2; median follow-up, 12.47 years). In Study 1, we studied the cross-sectional association between Lp(a) mass and MASLD stages (Analysis A). In Study 2, we studied the prospective association between Lp(a) concentration and MASLD, liver cirrhosis, and hepatocellular carcinoma (Analysis B). Finally, these analyses were accompanied by a prospective analysis using LPA Genetic Risk Score (Analysis C). RESULTS In Study 1, an inverse association between Lp(a) and at-risk metabolic dysfunction-associated steatohepatitis (odds ratioper 1-SD increase, 0.64; 95% confidence interval [CI], 0.42-0.97) was observed. In contrast, when similar associations were examined prospectively (Study 2), subjects with Lp(a) <10.75 nmol/L had a higher risk for cirrhosis (hazard ratio, 1.49; 95% CI, 1.22-1.81) and HCC (hazard ratio, 1.69; 95% CI, 1.12-2.56) compared with subjects with Lp(a) within the 10.75 to 21.5 nmol/L range. Above these levels, the risk increased significantly and positively. Similarly, genetic analysis showed an L-shaped association with LPA Genetic Risk Score. CONCLUSIONS The inverse association observed in cross-sectional studies should be attributed to reverse causality (ie, the presence of liver disease may decrease Lp(a) levels). Genetically predicted very low Lp(a) levels are also associated with impaired liver health prospectively. Clinical trials evaluating Lp(a)-lowering agents should thus be monitored carefully for adverse liver effects in subjects attaining extremely low concentrations.
Collapse
Affiliation(s)
- Hong Fan
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Matina Kouvari
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | | | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital, Westmead, New South Wales, Australia; University of Sydney, Sydney, New South Wales, Australia
| | - Georgios Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko," Athens, Greece
| | - Laura Valenzuela-Vallejo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Zhenqiu Liu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Xingdong Chen
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; Section of Endocrinology, VA Boston Healthcare System, Jamaica Plain, Massachusetts.
| |
Collapse
|
15
|
Li P, Liu X, Wu L, Dong L, Zhou J, Song Z. Thyroid Function and Brain Structure: Insight from a Mendelian Randomization Study. Neuroendocrinology 2024; 115:60-71. [PMID: 39653027 DOI: 10.1159/000542955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
INTRODUCTION Thyroid hormones play a critical role in brain development. However, the precise causal associations between thyroid function and structural changes in specific brain regions remain uncertain. METHODS We applied the univariate Mendelian randomization (UVMR) method to assess the causal effects of thyroid function on brain structure. Genome-wide association study (GWAS) data on thyroid-related traits from the ThyroidOmics Consortium including free thyroxine (FT4), free tri-iodothyronine (FT3), thyroid-stimulating hormone (TSH), FT3/FT4 ratio, as well as dichotomized high and low TSH levels were used as exposures. GWAS data on cortical thickness, surface area, and volume of subcortical structures served as outcomes. Inverse variance weighted was the main estimate method. Subsequently, multivariable MR (MVMR) was conducted to validate significant causal associations identified in UVMR. RESULTS UVMR analysis demonstrated a statistically significant inverse association between genetically predicted FT4 and putamen volume (β = -71.91 mm3, 95% confidence interval: -112.11 mm3 to -31.71 mm3, p = 4.54 × 10-4). The findings were robust in sensitivity analysis. MVMR analysis further confirmed a persistent causal relationship between FT4 and putamen volume after adjusting for FT3, TSH, and neuropsychiatric disorders. Functional enrichment analyses indicated the pathways by which FT4 influences putamen volume may be related to the thyroid hormone signaling pathway, sodium-independent organic anion transport, and Rap1 signaling pathway. CONCLUSION MR analysis provides evidence for causal relationships between thyroid function and brain structural alterations, particularly highlighting the impact of FT4 on putamen volume. Further research is warranted to elucidate the underlying mechanisms by which thyroid hormones modulate brain structure.
Collapse
Affiliation(s)
- Ping Li
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China,
| | - Xiao Liu
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Liming Wu
- Department of Urology, Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, China
| | - Liming Dong
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jianbo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhihui Song
- Department of Pharmacy, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Melini S, Pirozzi C, Lama A, Comella F, Opallo N, Del Piano F, Di Napoli E, Mollica MP, Paciello O, Ferrante MC, Mattace Raso G, Meli R. Co-Micronized Palmitoylethanolamide and Rutin Associated With Hydroxytyrosol Recover Diabesity-Induced Hepatic Dysfunction in Mice: In Vitro Insights Into the Synergistic Effect. Phytother Res 2024; 38:6035-6047. [PMID: 39474783 PMCID: PMC11634826 DOI: 10.1002/ptr.8361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/05/2024] [Accepted: 10/04/2024] [Indexed: 12/13/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) and diabesity (diabetes related to obesity) are interrelated since glucose and lipid alterations play a vital role in the development of both disorders. Due to their multi-variant metabolic features, more than one drug or natural product may be required to achieve proper therapeutic effects. This study aimed to evaluate the effectiveness of a formulation containing co-micronized palmitoylethanolamide and rutin (PEA-Rut) associated with hydroxytyrosol (HT), namely NORM3, against hepatic damage and metabolic alterations in high-fat diet (HFD)-induced diabesity in mice. NORM3 decreased the body weight and fat mass of obese mice. The formulation improved HFD-altered insulin sensitivity and hepatic glucose production and metabolism, as shown by glucose, insulin, pyruvate tolerance tests, Western blot, and real-time PCR. In the liver, NORM3 limited macro- and micro-vacuolar steatosis, as revealed by morphological analysis, and reduced the associated hepatic inflammation. NORM3 counteracted lipid dysfunctions of HFD animals, activating AMPK, a key cellular energy sensor, and normalizing the expression of carnitine palmitoyl-transferase (CPT)1, a rate-limiting enzyme of fatty acid β-oxidation, and other genes involved in lipid homeostasis. Relevantly, the hepatic antioxidant activity of NORM3 was proved (reduced ROS and increased detoxifying factors and enzymes). Finally, in vitro synergistic protective effects of the components (PEA-Rut and HT) on H2O2-induced oxidative challenge in HepG2 were determined (ROS production, inflammation, and antioxidant defense). Our results show the beneficial effect of NORM3 and its potential as an innovative phytotherapeutic combination in limiting hepatic damage progression and counteracting glucose and lipid dysmetabolism associated with diabesity.
Collapse
Affiliation(s)
- S. Melini
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - C. Pirozzi
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - A. Lama
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - F. Comella
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - N. Opallo
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - F. Del Piano
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - E. Di Napoli
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - M. P. Mollica
- Department of BiologyUniversity of Naples Federico IINaplesItaly
| | - O. Paciello
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - M. C. Ferrante
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - G. Mattace Raso
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - R. Meli
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
17
|
Pan C, Yu T, Zhao H, He J, Lu X, Tang H, Hong Y, Shang C, Wu Q, Yang A, Li C, Zhou M, Shi Y. Evaluation of pancreatic iodine uptake and related influential factors in multiphase dual-energy CT. Eur Radiol 2024; 34:7609-7621. [PMID: 38913243 DOI: 10.1007/s00330-024-10850-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/25/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024]
Abstract
OBJECTIVES To establish normative values and identify potential factors influencing pancreatic iodine uptake using dual-energy CT (DECT). MATERIALS AND METHODS This retrospective study included participants without pancreatic diseases undergoing DECT at two institutions with different platforms. Their protocols both included arterial phase (AP), portal venous phase (PP), and equilibrium phase (EP), defined as 35 s-40 s, 60 s-70 s, and 150 s-180 s after injection of contrast agent, respectively. Both iodine concentration (IC) and normalised IC (NIC) were measured. Demographic features, local measurements of the pancreas and visceral fat area (VFA) were considered as potential factors influencing iodine uptake using multivariate linear regression analyses. RESULTS A total of 562 participants (median age 58 years [interquartile range: 47-67], with 282 men) were evaluated. The mean IC differed significantly between two institutions (all p < 0.001) across three contrast-enhanced phases, while the mean NIC showed no significant differences (all p > 0.05). The mean values of NIC were 0.22 at AP, 0.43 at PP and 0.45 at EP. NICAP was independently affected by VFA (β = 0.362, p < 0.001), smoking (β = -0.240, p = 0.001), and type-II diabetes (β = -0.449, p < 0.001); NICPP by VFA (β = -0.301, p = 0.017) and smoking (β = -0.291, p < 0.001); and NICEP by smoking (β = -0.154, p = 0.10) and alcohol consumption (β = -0.350, p < 0.001) with statistical power values over 0.81. CONCLUSION NIC values were consistent across institutions. Abdominal obesity, smoking, alcohol consumption, and diabetes are independent factors influencing pancreatic iodine uptake. CLINICAL RELEVANCE STATEMENT This study has provided reference normative values, influential factors and effective normalisation methods of pancreatic iodine uptake in multiphase dual-energy CT for future studies in this area as a new biological marker. KEY POINTS Evaluation of pancreatic iodine uptake measured by dual-energy CT is a promising method for future studies. Abdominal obesity, smoking, alcohol consumption, diabetes, and sex are independent factors influencing pancreatic iodine uptake. Utility of normalised iodine concentration is necessary to ensure the consistency across different institutions.
Collapse
Affiliation(s)
- Chen Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Yu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jiani He
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xiaomei Lu
- CT Clinical Science CT, Philips Healthcare, Shenyang, China
| | - Haiyan Tang
- Department of Radiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yang Hong
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, China
| | - Chao Shang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Qijun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Aoran Yang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunli Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minghui Zhou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yu Shi
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
18
|
Yu C, Ding C, Yu C, Bao H, Cheng X. Decoding the fatty liver-hyperuricemia link in the obese and nonobese hypertensive patients: insights from a cohort study. Sci Rep 2024; 14:29525. [PMID: 39604465 PMCID: PMC11603371 DOI: 10.1038/s41598-024-80895-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic-dysfunction-associated fatty liver disease (MAFLD) and serum uric acid are closely related to cardiovascular and cerebrovascular diseases. However, the causal association between MAFLD and serum uric acid remains unclear. A total of 3417 patients without hyperuricemia were included in the final analysis. MAFLD was defined as fatty liver index (FLI) ≥ 30. Multivariate Cox regression analysis was used to explore the association between FLI and new-onset hyperuricemia. Restricted cubic splines and threshold saturation effect analysis were used to detect nonlinear associations. The mean age was 62.8 ± 8.3 year, and 68.5% were women. A total of 738 (21.6%) hypertensive patients developed new-onset hyperuricemia, 388 (11.4%) new-onset hyperuricemia10 and 190 (5.6%) new-onset hyperuricemia20 during the 4-year midday follow-up period. In the fully adjusted model, compared with the Q1 (FLI ≤ 8.5) group, the risk of hyperuricemia increased by 56% (HR: 1.56; 95% CI: 1.02, 2.38) in the Q4 (FLI > 39.4) group, new-onset hyperuricemia10 increased by 108% (HR: 2.08; 95% CI: 1.15, 3.78), and new-onset hyperuricemia20 increased by 156% (HR: 2.56; 95% CI: 1.11, 5.94), respectively. Saturation effects showed a nonlinear association between FLI and new-onset hyperuricemia (p for log likelihood ratio test < 0.05). Subgroup analysis and stratified analysis showed that there had a significantly higher risk of new-onset hyperuricemia in the patients with normal body mass index (< 24 kg/m2) (p for interaction: 0.018) and non-central obesity (p for interaction: 0.024). MAFLD is an independent risk factor for hyperuricemia in hypertensive patients, especially in patients with normal body mass index and non-central obesity.
Collapse
Affiliation(s)
- Chuanli Yu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| | - Congcong Ding
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| | - Chao Yu
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| | - Huihui Bao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China.
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China.
| | - Xiaoshu Cheng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Jiang F, Wang L, Ying H, Sun J, Zhao J, Lu Y, Bian Z, Chen J, Fang A, Zhang X, Larsson SC, Mantzoros CS, Wang W, Yuan S, Ding Y, Li X. Multisystem health comorbidity networks of metabolic dysfunction-associated steatotic liver disease. MED 2024; 5:1413-1423.e3. [PMID: 39116870 DOI: 10.1016/j.medj.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/09/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND The global burden of metabolic dysfunction-associated steatotic liver disease (MASLD) is growing, but its subsequent health consequences have not been thoroughly examined. METHODS A phenome-wide association study was conducted to map the associations of MASLD with 948 unique clinical outcomes among 361,021 Europeans in the UK Biobank. Disease trajectory and comorbidity analyses were applied to visualize the sequential patterns of multiple comorbidities related to the occurrence of MASLD. The associations jointly verified by observational and polygenic phenome-wide analyses were further replicated by two-sample Mendelian randomization analysis using data from the FinnGen study and international consortia. FINDINGS The observational and polygenic phenome-wide association study revealed the associations of MASLD with 96 intrahepatic and extrahepatic diseases, including circulatory, metabolic, genitourinary, neurological, gastrointestinal, and hematologic diseases. Sequential patterns of MASLD-related extrahepatic comorbidities were primarily found in circulatory, metabolic, and inflammatory diseases. Mendelian randomization analyses supported the causal associations between MASLD and the risk of several intrahepatic disorders, metabolic diseases, cardio-cerebrovascular disease, and ascites but found no associations with neurological diseases. CONCLUSIONS This study elucidated multisystem comorbidities and health consequences of MASLD, contributing to the development of combination interventions targeting distinct pathways for health promotion among patients with MASLD. FUNDING X.L. was funded by the Natural Science Fund for Distinguished Young Scholars of Zhejiang Province (LR22H260001) and the National Nature Science Foundation of China (82204019) and Y.D. was funded by the Key Project of Traditional Chinese Medicine Science and Technology Plan of Zhejiang Province (GZY-ZJ-KJ-24077) and the National Natural Science Foundation of China (82001673 and 82272860).
Collapse
Affiliation(s)
- Fangyuan Jiang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Lijuan Wang
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China; Centre for Global Health, Usher Institute, the University of Edinburgh, Edinburgh, UK
| | - Haochao Ying
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Sun
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhui Zhao
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Lu
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Zilong Bian
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Chen
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Aiping Fang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Xuehong Zhang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Susanna C Larsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Unit of Medical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuai Yuan
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xue Li
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Li J, Yan N, Li X, He S. Association between serum vitamin D concentration and liver fibrosis in diabetes mellitus patients: a cross-sectional study from the NHANES database. Acta Diabetol 2024; 61:1393-1402. [PMID: 38831202 DOI: 10.1007/s00592-024-02292-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/14/2024] [Indexed: 06/05/2024]
Abstract
AIM Liver fibrosis (LF) is a common complication of diabetes mellitus (DM). Studies have found that vitamin D (VD), as a modifiable factor has been reported to be associated with LF. The relationship between serum VD concentration and LF in DM patients has rarely been reported. The aim of this study was to assess the association between serum VD concentration and LF in DM patients. METHODS In this cross-sectional study, data of DM patients aged ≥ 45 years were extracted from the National Health and Nutrition Examination Survey (NHANES 2017-2018). Serum VD concentration was measured by high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Vibration controlled transient elastography (VCTE) was used to measure liver stiffness. Covariates included sociodemographic information, lifestyles, laboratory data, diseases history were extracted from the database. The weighted univariable and multivariable logistic regression models were utilized to explore the association between serum VD concentration and LF in DM patients, and were described as odds ratio (ORs) and 95% confidence intervals (CIs). Subgroup analyses based on BMI, liver steatosis, hypertension and dyslipidemia were further assessed the association. RESULTS A total of 799 patients were included, of which 188 (23.53%) had LF. Higher serum VD concentration was associated with the lower odds of LF (OR = 0.33, 95% CI 0.19-0.59) and advanced LF (OR = 0.31, 95% CI 0.17-0.55) in DM patients after adjustment for race, liver steatosis, BMI, smoking, drinking, AST, ALT and physical activity, especially in patients with liver steatosis (OR = 0.28, 95% CI 0.13-0.59) and dyslipidemia (OR = 0.31, 95% CI 0.14-0.66), respectively. CONCLUSIONS High serum VD concentration may have a potential benefit for maintain the liver health in DM patients.
Collapse
Affiliation(s)
- Jing Li
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China
| | - Ni Yan
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China
| | - Xiaofeng Li
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China
| | - Shenglin He
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China.
| |
Collapse
|
21
|
Wang Y, Zhou J, Yang Q, Li X, Qiu Y, Zhang Y, Liu M, Zhu AJ. Therapeutic siRNA targeting PLIN2 ameliorates steatosis, inflammation, and fibrosis in steatotic liver disease models. J Lipid Res 2024; 65:100635. [PMID: 39187042 PMCID: PMC11440260 DOI: 10.1016/j.jlr.2024.100635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/10/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide. If left untreated, MASLD can progress from simple hepatic steatosis to metabolic dysfunction-associated steatohepatitis, which is characterized by inflammation and fibrosis. Current treatment options for MASLD remain limited, leaving substantial unmet medical needs for innovative therapeutic approaches. Here, we show that PLIN2, a lipid droplet protein inhibiting hepatic lipolysis, serves as a promising therapeutic target for MASLD. Hepatic PLIN2 levels were markedly elevated in multiple MASLD mouse models induced by diverse nutritional and genetic factors. The liver-specific deletion of Plin2 exhibited significant anti-MASLD effects in these models. To translate this discovery into a therapeutic application, we developed a GalNAc-siRNA conjugate with enhanced stabilization chemistry and validated its potent and sustained efficacy in suppressing Plin2 expression in mouse livers. This siRNA therapeutic, named GalNAc-siPlin2, was shown to be biosafe in mice. Treatment with GalNAc-siPlin2 for 6-8 weeks led to a decrease in hepatic triglyceride levels by approximately 60% in high-fat diet- and obesity-induced MASLD mouse models, accompanied with increased hepatic secretion of VLDL-triglyceride and enhanced thermogenesis in brown adipose tissues. Eight-week treatment with GalNAc-siPlin2 significantly improved hepatic steatosis, inflammation, and fibrosis in high-fat/high fructose-induced metabolic dysfunction-associated steatohepatitis models compared to control group. As a proof of concept, we developed a GalNAc-siRNA therapeutic targeting human PLIN2, which effectively suppressed hepatic PLIN2 expression and ameliorated MASLD in humanized PLIN2 knockin mice. Together, our results highlight the potential of GalNAc-siPLIN2 as a candidate MASLD therapeutic for clinical trials.
Collapse
Affiliation(s)
- Yao Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Jiaxin Zhou
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Qi Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Xinmeng Li
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yifu Qiu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yansong Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, China.
| | - Min Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | - Alan Jian Zhu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Kara YB, Ozel Y. Comparison of the Grades of Fatty Liver Disease With Perioperative Risk Factors in Patients With Laparoscopic Sleeve Gastrectomy. Cureus 2024; 16:e69717. [PMID: 39429298 PMCID: PMC11490200 DOI: 10.7759/cureus.69717] [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: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Background Obesity is a global healthcare problem, and nonalcoholic fatty liver disease (NAFLD) is a commonly observed comorbid disease in the bariatric population. This study evaluated the relationship between NAFLD and various risk factors, including demographic, biochemical, and comorbid conditions in patients undergoing laparoscopic sleeve gastrectomy (LSG). Material and methods This retrospective data analysis study included patients who underwent LSG between August 2023 and 2024. Patient demographic data were collected, such as age, gender, weight, and body mass index (BMI), and NAFLD grade was determined by ultrasonography. Biochemical markers were recorded, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol, triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), fasting blood glucose (FBG), HbA1c, and vitamin D. The presence of type II diabetes mellitus (T2DM) and hypertension (HT) was evaluated and compared with the grade of hepatosteatosis. Results The study included 436 patients, of whom 73.6% (n = 321) were female. The mean age was 37.23 ± 10.49 years, and the mean BMI value was 41.25 ± 6.11 kg/m2. Patients were classified and compared according to their NAFLD grade, revealing statistically significant differences in weight, BMI, ALT, AST, HDL, LDL, TG, total cholesterol, HbA1c, FBG, vitamin D, obesity class, DM, and HT (p < 0.05). HDL and vitamin D showed an inverse correlation with NAFLD. We observed no significant difference in the relationship of NAFLD with age and the presence of gallstone. Logistic regression analysis revealed that ALT, AST, LDL, total cholesterol, and FBG were statistically significantly associated with NAFLD in the multivariate model. Conclusion Hepatosteatosis, T2DM, and HT are frequent comorbid diseases that are common in bariatric patients. Our study shows that ALT, AST, LDL, FBG, and total cholesterol may be used as predictors of NAFLD.
Collapse
Affiliation(s)
- Yalçın Burak Kara
- Department of General Surgery, Bahcesehir University, School of Medicine, Istanbul, TUR
- Department of General Surgery, Medical Park Pendik Hospital, Istanbul, TUR
| | - Yahya Ozel
- General Surgery, Dogus University School of Medicine, Istanbul, TUR
- Department of General Surgery, Medical Park Pendik Hospital, Istanbul, TUR
| |
Collapse
|
23
|
Hu L, Wu X, Peng J, Yan B, Li J, Guo Y, Han J. Subchronic oral toxic effects of 2,4-dinitroaniline in wistar rats: A comprehensive toxicity evaluation. Food Chem Toxicol 2024; 191:114846. [PMID: 38960084 DOI: 10.1016/j.fct.2024.114846] [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: 04/03/2024] [Revised: 06/21/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
2,4-dinitroaniline (2,4-D), a widely used dye intermediate, is one of the typical pollutants, and its potential health risks and toxicity are still largely unknown. To explore its subchronic oral toxicity, Wistar rats (equal numbers of males and females) were used as test animals, and a 90-day oral dosing experiment was conducted, divided into control group, low-dose group (0.055 mg/kg), medium-dose group (0.22 mg/kg), medium-high dose group (0.89 mg/kg), and high-dose group (3.56 mg/kg). The body weight data, clinical appearance, and drug reactions of each test rat within 90 days of dosing were recorded; morning urine samples were collected four times to test for eight urinary indicators; blood samples were collected to test for nineteen hematological indicators and sixteen biochemical indicators; tissue samples were collected for pathological analysis; moreover, the no-observed-adverse-effect level (NOAEL) was determined, and the benchmark dose method was used to support this determination and provide a statistical estimate of the dose corresponding. The results indicated that the chronic toxicity of 2,4-dinitroaniline showed certain gender differences, with the eyes, liver, and kidneys being the main potential target organs of toxicity. Moreover, the subchronic oral NOAEL for 2,4-dinitroaniline was determined to be 0.22 mg/kg body weight (0.22 mg/kg for males and 0.89 mg/kg for females), and a preliminary calculation of the safe exposure limit for human was 0.136 mg/kg. The research results greatly enriched the safety evaluation data of 2,4-dinitroaniline, contributing to a robust scientific foundation for the development of informed safety regulations and public health precautions.
Collapse
Affiliation(s)
- Ling Hu
- Wuhan Science and Technology Center of Ecology and Environment, Wuhan 430015, China
| | - Xiaoxu Wu
- Wuhan Science and Technology Center of Ecology and Environment, Wuhan 430015, China
| | - Jinjin Peng
- Wuhan Science and Technology Center of Ecology and Environment, Wuhan 430015, China
| | - Biao Yan
- Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academic of Sciences, Wuhan 430072, China.
| | - Jinquan Li
- Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yongyong Guo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academic of Sciences, Wuhan 430072, China
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academic of Sciences, Wuhan 430072, China.
| |
Collapse
|
24
|
Zheng M, Xu J, Feng Z. Association between nonalcoholic fatty liver disease and bone mineral density: Mendelian randomization and mediation analysis. Bone Rep 2024; 22:101785. [PMID: 39220175 PMCID: PMC11363625 DOI: 10.1016/j.bonr.2024.101785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 09/04/2024] Open
Abstract
Background Observational studies have reported significant association between non-alcoholic fatty liver disease (NAFLD) and bone mineral density (BMD), a critical indicator of bone health. We aimed to investigate whether NAFLD is a cause for changes in BMD. Methods We selected 29 independent SNPs as instrumental variables for NAFLD. A range of Mendelian randomization (MR) methods, namely the inverse variance-weighted (IVW) method, weighted-median, weighted-mode, and MR-Egger regression, were utilized to determine the causal effects of NAFLD on BMD. Two-step MR analysis was conducted to determine the mediating effect of fasting glucose, insulin, glycosylated hemoglobin, low-density cholesterol, and body-mass index on the association between NAFLD and BMD. False-discovery-rate (FDR) was used to correct for multiple testing bias. Results The IVW-method indicated a significantly inverse association between genetically predicted NAFLD and total body BMD (β = -0.04, 95 % CI -0.07 to -0.02, FDR = 0.010). Notably, the relationship was more pronounced in participants over 60 years of age (β = -0.06, 95 % CI -0.11 to -0.02, FDR = 0.030). Inverse associations were observed in other subpopulations and in site-specific BMD, though they were not statistically significant after correcting for multiple testing. We observed a significantly positive association between NAFLD and the risk of osteoporosis. Consistency in results was observed across multiple MR methods and in the repeated analysis. Fasting glucose, insulin, and glycosylated hemoglobin mediated 25.4 % (95 % CI 17.6-31.5 %), 18.9 % (12.0-24.9 %), and 27.9 % (19.9-36.7 %) of the effect of NAFLD on BMD, respectively. Conclusion Our findings underscore a probable causal negative link between NAFLD and BMD, indicating that NAFLD might detrimentally affect bone health, especially in older individuals.
Collapse
Affiliation(s)
- Minzhe Zheng
- Department of Orthopedics, the Affiliated Lihuili Hospital, Ningbo University, Ningbo City, China
| | - Junxiang Xu
- Department of Orthopedics, the Affiliated Lihuili Hospital, Ningbo University, Ningbo City, China
| | - Zongxian Feng
- Department of Orthopedics, the Affiliated Lihuili Hospital, Ningbo University, Ningbo City, China
| |
Collapse
|
25
|
Yuan W, Ran Y, Wang J, Pei F, Cui L, Chen S, Wu S, Zhou L. Mediating effect of diabetes on the relationship between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease: a prospective cohort study. Eur J Gastroenterol Hepatol 2024; 36:1133-1140. [PMID: 39101442 DOI: 10.1097/meg.0000000000002794] [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] [Indexed: 08/06/2024]
Abstract
OBJECTIVE This study explored the mediating effect of diabetes on the relationship between nonalcoholic fatty liver disease (NAFLD) and atherosclerotic cardiovascular disease (ASCVD). METHODS In this prospective community cohort study, 82 975 participants were enrolled, with the primary outcome being the incidence of new-onset ASCVD. Using the Cox proportional hazards model, the hazard ratio (HR) and 95% confidence interval (CI) for ASCVD occurrence were computed between NAFLD and non-NAFLD groups. The correlation between NAFLD and diabetes was assessed using a binary logistic regression model, and that between NAFLD, diabetes and ASCVD using a mediation model. RESULTS During follow-up, 9471 ASCVD cases were observed. Compared with individuals without NAFLD, those with NAFLD showed an increased ASCVD risk (HR: 1.424; 95% CI: 1.363-1.488; P < 0.001). Stratifying NAFLD based on metabolic subphenotypes revealed a higher ASCVD risk in the NAFLD combined with diabetes subgroup than in the non-NAFLD subgroup (HR: 1.960; 95% CI: 1.817-2.115; P < 0.001). NAFLD was positively associated with baseline diabetes (odds ratio: 2.983; 95% CI: 2.813-3.163; P < 0.001). Furthermore, NAFLD severity was positively correlated with diabetes risk. Mediation analysis indicated that diabetes partially mediated the effect of NAFLD on ASCVD incidence, accounting for 20.33% of the total effect. CONCLUSION NAFLD is an independent predictor of increased ASCVD risk, which may be slightly mediated by diabetes in patients with NAFLD. Evaluating NAFLD and diabetes may be crucial in the early screening and prevention of ASCVD.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
- Rheumatology and Immunology
| | - Ying Ran
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
| | | | | | | | - Shuohua Chen
- Cardiology, Kailuan General Hospital, Tangshan, China
| | - Shouling Wu
- Cardiology, Kailuan General Hospital, Tangshan, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
| |
Collapse
|
26
|
Luo Q, Liu G, Li Q, Lu J, Zheng W, Huang Y, Li C. Novel insights into causal effects of maternal nonalcoholic fatty liver disease on adverse pregnancy outcomes: evidence from Human Genetics and Mendelian Randomization Study. Eur J Clin Nutr 2024:10.1038/s41430-024-01489-7. [PMID: 39147848 DOI: 10.1038/s41430-024-01489-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Observational studies have associated nonalcoholic fatty liver disease (NAFLD) with adverse pregnancy events, but findings show heterogeneity, leaving the causal direction and mediating pathways unclear. We aimed to investigate the causal relation between NAFLD and various pregnancy events, and to elucidate the underlying mediating pathways while determining the proportion of this correlation that is mediated through these pathways. METHODS A genome-wide association study involving over 6 million participants employing Mendelian randomization (MR) and mediation analysis was performed. The study used genetically predicted NAFLD as exposures and cardiometabolic traits as mediators, with various adverse pregnancy events as outcomes. The main analysis was performed using the inverse variance weighted (IVW) approach, while sensitivity analyses included the weighted median, weighted mode, MR-Egger, and MR-PRESSO methods. Mediation analyses were performed using a two-step MR framework. RESULTS In this MR cohort study, NAFLD was found to be strongly associated with elevated risks of GDM (P = 0.019 for the discovery dataset, P < 0.001 for the discovery dataset) and HDPs, including any HDP (P < 0.001 for the both datasets), gestational hypertension (P = 0.007 for the discovery dataset, P < 0.001 for the discovery dataset), and pre-eclampsia or eclampsia (P = 0.040 for the discovery dataset, P < 0.001 for the discovery dataset). However, no significant associations were found with hemorrhage in early pregnancy, postpartum hemorrhage, preterm birth, or offspring birthweight for both datasets. Cardiometabolic traits played a significant mediating role in these associations, rather than solely acting as confounding factors. CONCLUSIONS This study provided evidence supporting a correlation between NAFLD and a higher risk of adverse pregnancy events and introduces some new insights. These findings may inform preventions and interventions for remediating adverse pregnancy outcomes attributable to NAFLD.
Collapse
Affiliation(s)
- Qiuyan Luo
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Guoting Liu
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Qiulan Li
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Jinghong Lu
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Wenjing Zheng
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Yukui Huang
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China.
| | - Cun Li
- Department of Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, PR China.
| |
Collapse
|
27
|
Chen C, Yang K, Zhang Y, Lu M, Zhao X, Wan Z. Pathogenic gene connections in type 2 diabetes and non-alcoholic fatty liver disease: a bioinformatics analysis and mouse model investigations experiments. Nutr Diabetes 2024; 14:60. [PMID: 39107295 PMCID: PMC11303809 DOI: 10.1038/s41387-024-00323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD) are prevalent metabolic disorders with overlapping pathophysiological mechanisms. A comprehensive understanding of the shared molecular pathways involved in these conditions can advance the development of effective therapeutic interventions. METHODS We used two datasets sourced from the Gene Expression Omnibus (GEO) database to identify common differentially expressed genes (DEGs) between T2D and NAFLD. Subsequently, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to identify the enriched biological processes and signaling pathways. In addition, we performed a protein-protein interaction (PPI) network analysis to identify hub genes with pivotal roles. To validate our findings, we established a type 2 diabetic mouse model with NAFLD. RESULTS Our analysis identified 53 DEGs shared between T2D and NAFLD. Enrichment analysis revealed their involvement in signal transduction, transcriptional regulation, and cell proliferation as well as in the ferroptosis signaling pathways. PPI network analysis identified ten hub genes, namely CD44, CASP3, FYN, KLF4, HNRNPM, HNRNPU, FUBP1, RUNX1, NOTCH3, and ANXA2. We validated the differential expression of FYN, HNRNPU, and FUBP1 in liver tissues of a type 2 diabetic mouse model with NAFLD. CONCLUSIONS Our study offers valuable insights into the shared molecular mechanisms underlying T2D and NAFLD. The identified hub genes and pathways present promising prospects as therapeutic targets to address these prevalent metabolic disorders.
Collapse
Affiliation(s)
- Chao Chen
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen, 361021, China.
| | - Kunhuan Yang
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yuhan Zhang
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Meiqi Lu
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xiaoyan Zhao
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Zheng Wan
- School of Medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
28
|
Drake I, Giontella A, Miari M, Önnerhag K, Orho-Melander M. Lifestyle and genetic risk of chronic liver disease in metabolically healthy and unhealthy individuals from the general population. JHEP Rep 2024; 6:101105. [PMID: 39049959 PMCID: PMC11268350 DOI: 10.1016/j.jhepr.2024.101105] [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: 11/20/2023] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 07/27/2024] Open
Abstract
Background & Aims It is unclear to what extent lifestyle and genetic factors affect the incidence of chronic liver disease (CLD) in the general population and if lifestyle affects CLD independently of underlying cardiometabolic perturbations and genetic predisposition. Methods We examined 27,991 men and women aged 44-73 years from the Malmö Diet and Cancer Study recruited between 1991-1996 and followed until the end of 2020 using registry linkage (median follow-up time 25.1 years; 382 incident first-time CLD events). Associations between cardiometabolic factors, polygenic risk scores (PRSs), and lifestyle factors in relation to CLD were examined using multivariable Cox proportional hazards regression models. Results The incidence of CLD increased with number of cardiometabolic risk factors (the hazard ratio per each additional cardiometabolic risk factor was 1.33; 95% CI 1.21-1.45; p = 5.1 x 10-10). Two novel PRSs for metabolic dysfunction-associated steatotic liver disease and a PRS for cirrhosis were associated with higher risk of CLD but provided marginal predictive utility on top of other risk factors and compared to the PNPLA3 rs738409 genetic variant. An unhealthy lifestyle (high alcohol intake, current smoking, physical inactivity and unhealthy diet) markedly increased the risk of CLD (hazard ratio 3.97, 95% CI 2.59-6.10). Observed associations between examined lifestyle factors and CLD were largely independent of cardiometabolic perturbations and polygenic risk. Conclusions We confirmed the importance of cardiometabolic dysfunction in relation to risk of CLD in the general population. Lifestyle risk factors were shown to be independently associated with CLD and added predictive information on top of cardiometabolic risk factors. Information on the polygenic risk of liver disease does not currently improve the prediction of CLD in the general population. Impact and implications This large population-based prospective study suggests largely independent roles of cardiometabolic, lifestyle, and genetic risk factors in the development of chronic liver disease. Findings strengthen the evidence base for a beneficial effect of modification of high-risk lifestyle behaviors in the primary prevention of chronic liver disease in the general population.
Collapse
Affiliation(s)
- Isabel Drake
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Skåne University Hospital, Malmö, Sweden
| | - Alice Giontella
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Mariam Miari
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Kristina Önnerhag
- Gastroenterology Research Unit, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- Department of Surgery and Gastroenterology, Skåne University Hospital, Malmö, Sweden
| | | |
Collapse
|
29
|
Liu Y, Wang Y, Xing Y, Wolters M, Shi D, Zhang P, Dang J, Chen Z, Cai S, Wang Y, Liu J, Wang X, Zhou H, Xu M, Guo L, Li Y, Song J, Li J, Dong Y, Cui Y, Hu P, Hebestreit A, Wang HJ, Li L, Ma J, Yeo YH, Wang H, Song Y. Establish a noninvasive model to screen metabolic dysfunction-associated steatotic liver disease in children aged 6-14 years in China and its applications in high-obesity-risk countries and regions. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 49:101150. [PMID: 39171077 PMCID: PMC11338159 DOI: 10.1016/j.lanwpc.2024.101150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/27/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024]
Abstract
Background The prevalence of metabolic-associated steatotic liver disease (MASLD) is rising precipitously among children, particularly in regions or countries burdened with high prevalence of obesity. However, identifying those at high risk remains a significant challenge, as the majority do not exhibit distinct symptoms of MASLD. There is an urgent need for a widely accepted non-invasive predictor to facilitate early disease diagnosis and management of the disease. Our study aims to 1) evaluate and compare existing predictors of MASLD, and 2) develop a practical screening strategy for children, tailored to local prevalence of obesity. Methods We utilized a school-based cross-sectional survey in Beijing as the training dataset to establish predictive models for screening MASLD in children. An independent school-based study in Ningbo was used to validate the models. We selected the optimal non-invasive MASLD predictor by comparing logistic regression model, random forest model, decision tree model, and support vector machine model using both the Beijing and Ningbo datasets. This was followed by serial testing using the best performance index we identified and indices from previous studies. Finally, we calculated the potential MASLD screening recommendation categories and corresponding profits based on national and subnational obesity prevalence, and applied those three categories to 200 countries according to their obesity prevalence from 1990 to 2022. Findings A total of 1018 children were included (NBeijing = 596, NNingbo = 422). The logistic regression model demonstrated the best performance, identifying the waist-to-height ratio (WHtR, cutoff value ≥0.48) as the optimal noninvasive index for predicting MASLD, with strong performance in both training and validation set. Additionally, the combination of WHtR and lipid accumulation product (LAP) was selected as an optimal serial test to improve the positive predictive value, with a LAP cutoff value of ≥668.22 cm × mg/dL. Based on the obesity prevalence among 30 provinces, three MASLD screening recommendations were proposed: 1) "Population-screening-recommended": For regions with an obesity prevalence ≥12.0%, where MASLD prevalence ranged from 5.0% to 21.5%; 2) "Resources-permitted": For regions with an obesity prevalence between 8.4% and 12.0%, where MASLD prevalence ranged from 2.3% to 4.4%; 3) "Population-screening-not-recommended": For regions with an obesity prevalence <8.4%, where MASLD prevalence is difficult to detect using our tool. Using our proposed cutoff for screening MASLD, the number of countries classified into the "Population-screening-recommended" and "Resources-permitted" categories increased from one and 11 in 1990 to 95 and 28 in 2022, respectively. Interpretation WHtR might serve as a practical and accessible index for predicting pediatric MASLD. A WHtR value ≥0.48 could facilitate early identification and management of MASLD in areas with obesity prevalence ≥12.0%. Furthermore, combining WHtR ≥0.48 with LAP ≥668.22 cm × mg/dL is recommended for individual MASLD screening. Moreover, linking these measures with population obesity prevalence not only helps estimate MASLD prevalence but also indicates potential screening profits in regions at varying levels of obesity risk. Funding This study was supported by grants from Capital's Funds for Health Improvement and Research (Grant No. 2022-1G-4251), National Natural Science Foundation of China (Grant No. 82273654), Major Science and Technology Projects for Health of Zhejiang Province (Grant No. WKJ-ZJ-2216), Cyrus Tang Foundation for Young Scholar 2022 (2022-B126) and Sino-German Mobility Programme (M-0015).
Collapse
Affiliation(s)
- Yunfei Liu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Youxin Wang
- Department of Maternal and Child Health, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yunfei Xing
- Department of Maternal and Child Health, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Maike Wolters
- Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology – BIPS, Bremen, Germany
| | - Di Shi
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Pingping Zhang
- Ningbo Center for Healthy Lifestyle Research, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China
| | - Jiajia Dang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Ziyue Chen
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Shan Cai
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yaqi Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Jieyu Liu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Xinxin Wang
- Linyi University, Linyi, Shandong Province, China
| | - Haoyu Zhou
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Miao Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China
| | - Lipo Guo
- Changping Health Education Center for Primary and Secondary Schools, Beijing, China
| | - Yuanyuan Li
- Changping Health Education Center for Primary and Secondary Schools, Beijing, China
| | - Jieyun Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Jing Li
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yanhui Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yanchun Cui
- Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Peijin Hu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Antje Hebestreit
- Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology – BIPS, Bremen, Germany
| | - Hai-Jun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Li Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang Province, China
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yee Hui Yeo
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Hui Wang
- Department of Maternal and Child Health, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yi Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| |
Collapse
|
30
|
Lee SHT, Garske KM, Arasu UT, Kar A, Miao Z, Alvarez M, Koka A, Darci-Maher N, Benhammou JN, Pan DZ, Örd T, Kaminska D, Männistö V, Heinonen S, Wabitsch M, Laakso M, Agopian VG, Pisegna JR, Pietiläinen KH, Pihlajamäki J, Kaikkonen MU, Pajukanta P. Single nucleus RNA-sequencing integrated into risk variant colocalization discovers 17 cell-type-specific abdominal obesity genes for metabolic dysfunction-associated steatotic liver disease. EBioMedicine 2024; 106:105232. [PMID: 38991381 PMCID: PMC11663762 DOI: 10.1016/j.ebiom.2024.105232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Abdominal obesity increases the risk for non-alcoholic fatty liver disease (NAFLD), now known as metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS To elucidate the directional cell-type level biological mechanisms underlying the association between abdominal obesity and MASLD, we integrated adipose and liver single nucleus RNA-sequencing and bulk cis-expression quantitative trait locus (eQTL) data with the UK Biobank genome-wide association study (GWAS) data using colocalization. Then we used colocalized cis-eQTL variants as instrumental variables in Mendelian randomization (MR) analyses, followed by functional validation experiments on the target genes of the cis-eQTL variants. FINDINGS We identified 17 colocalized abdominal obesity GWAS variants, regulating 17 adipose cell-type marker genes. Incorporating these 17 variants into MR discovers a putative tissue-of-origin, cell-type-aware causal effect of abdominal obesity on MASLD consistently with multiple MR methods without significant evidence for pleiotropy or heterogeneity. Single cell data confirm the adipocyte-enriched mean expression of the 17 genes. Our cellular experiments across human adipogenesis identify risk variant -specific epigenetic and transcriptional mechanisms. Knocking down two of the 17 genes, PPP2R5A and SH3PXD2B, shows a marked decrease in adipocyte lipidation and significantly alters adipocyte function and adipogenesis regulator genes, including DGAT2, LPL, ADIPOQ, PPARG, and SREBF1. Furthermore, the 17 genes capture a characteristic MASLD expression signature in subcutaneous adipose tissue. INTERPRETATION Overall, we discover a significant cell-type level effect of abdominal obesity on MASLD and trace its biological effect to adipogenesis. FUNDING NIH grants R01HG010505, R01DK132775, and R01HL170604; the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (Grant No. 802825), Academy of Finland (Grants Nos. 333021), the Finnish Foundation for Cardiovascular Research the Sigrid Jusélius Foundation and the Jane and Aatos Erkko Foundation; American Association for the Study of Liver Diseases (AASLD) Advanced Transplant Hepatology award and NIH/NIDDK (P30DK41301) Pilot and Feasibility award; NIH/NIEHS F32 award (F32ES034668); Finnish Diabetes Research Foundation, Kuopio University Hospital Project grant (EVO/VTR grants 2005-2021), the Academy of Finland grant (Contract no. 138006); Academy of Finland (Grant Nos 335443, 314383, 272376 and 266286), Sigrid Jusélius Foundation, Finnish Medical Foundation, Finnish Diabetes Research Foundation, Novo Nordisk Foundation (#NNF20OC0060547, NNF17OC0027232, NNF10OC1013354) and Government Research Funds to Helsinki University Hospital; Orion Research Foundation, Maud Kuistila Foundation, Finish Medical Foundation, and University of Helsinki.
Collapse
Affiliation(s)
- Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Uma Thanigai Arasu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nicholas Darci-Maher
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jihane N Benhammou
- Vatche and Tamar Manoukian Division of Digestive Diseases and Gastroenterology, Hepatology and Parenteral Nutrition, David Geffen School of Medicine at UCLA and VA Greater Los Angeles HCS, Los Angeles, CA, USA
| | - David Z Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Tiit Örd
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Division of Cardiology, Department of Medicine, UCLA, Los Angeles, CA, USA
| | - Ville Männistö
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland; Department of Internal Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Vatche G Agopian
- Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joseph R Pisegna
- Department of Medicine and Human Genetics, Division of Gastroenterology, Hepatology and Parenteral Nutrition, David Geffen School of Medicine at UCLA and VA Greater Los Angeles HCS, Los Angeles, CA, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Healthy WeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Minna U Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Liu Y, Liang X, Hu Y, Zhang N, Zhu X, Feng Y, Qin Z, Wang Z, Kangzhuo B, Xiao X, Zhao X. Temporal relationship between hepatic steatosis and fasting blood glucose elevation: a longitudinal analysis from China and UK. BMC Public Health 2024; 24:1865. [PMID: 38997689 PMCID: PMC11241918 DOI: 10.1186/s12889-024-19177-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND The link between nonalcoholic fatty liver disease and type 2 diabetes has not been fully established. We investigated the temporal relationship between nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), quantitatively assessed the impact, and evaluated the related mediation effect. METHODS This study involved participants from the China Multi-Ethnic Cohort Study and the UK Biobank. We performed cross-lagged path analysis to compare the relative magnitude of the effects between NAFLD and T2D using two-period biochemical data. Hepatic steatosis and fasting blood glucose elevation (FBG) represented NAFLD and T2D respectively. We fitted two separate Cox proportional-hazards models to evaluate the influence of hepatic steatosis on T2D. Furthermore, we applied the difference method to assess mediation effects. RESULTS In cross-lagged path analyses, the path coefficients from baseline hepatic steatosis to first repeat FBG (βCMEC = 0.068, βUK-Biobank = 0.033) were significantly greater than the path coefficients from baseline FBG to first repeat hepatic steatosis (βCMEC = 0.027, βUK-Biobank = -0.01). Individuals with hepatic steatosis have a risk of T2D that is roughly three times higher than those without the condition (HR = 3.478 [3.314, 3.650]). Hepatic steatosis mediated approximately 69.514% of the total effect between obesity and follow-up T2D. CONCLUSIONS Our findings contribute to determining the sequential relationship between NAFLD and T2D in the causal pathway, highlighting that the dominant pathway in the relationship between these two early stages of diseases was the one from hepatic steatosis to fasting blood glucose elevation. Individuals having NAFLD face a significantly increased risk of T2D and require long-term monitoring of their glucose status as well.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CN, 610041, China
| | - Xian Liang
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Yifan Hu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CN, 610041, China
| | - Ning Zhang
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CN, 610041, China
| | - Xingren Zhu
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CN, 610041, China
| | - Yuemei Feng
- School of Public Health, Kunming Medical University, Kunming, China
| | - Zixiu Qin
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Zihao Wang
- Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Baima Kangzhuo
- High Altitude Health Science Research Center of Tibet University, Lhasa, Tibet, China
| | - Xiong Xiao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CN, 610041, China.
| | - Xing Zhao
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, CN, 610041, China
| |
Collapse
|
32
|
Behari J, Wang R, Luu HN, McKenzie D, Molinari M, Yuan JM. Severe obesity is associated with worse outcomes than lean metabolic dysfunction-associated steatotic liver disease. Hepatol Commun 2024; 8:e0471. [PMID: 38934706 PMCID: PMC11213590 DOI: 10.1097/hc9.0000000000000471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/25/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is highly prevalent in people with obesity. We aimed to study the association of body mass index (BMI) with clinical outcomes in patients with MASLD. METHODS A retrospective cohort of 32,900 patients with MASLD, identified through the International Classification of Diseases-9 and 10 codes within the electronic health records of a large US-based health system, with a mean follow-up of 5.5 years (range: 1-15 y), was stratified into 6 BMI categories, <25, 25-<30, 30-<40, 40-<50, and ≥50 kg/m2. RESULTS The risk of liver decompensation and extrahepatic obesity-associated cancers had a J-shaped profile (both ps for linear and quadratic terms <0.05). Compared to patients with BMI 25-<30 kg/m2, the adjusted HRs (95% CIs) for liver decompensation of patients with BMI <25 and BMI ≥50 kg/m2 were 1.44 (1.17-1.77) and 2.27 (1.66-3.00), respectively. The corresponding figures for obesity-associated extrahepatic cancer were 1.15 (0.97-1.36) and 1.29 (1.00-1.76). There was an inverse association for BMI with liver transplantation and non-obesity-associated cancer (both ps for linear terms <0.05), but no association with HCC or all types of cancers combined. A similar J-shaped association between BMI and all-cause mortality was observed; adjusted HRs (95% CIs) for BMI <25 and ≥50 kg/m2 were 1.51 (1.32-1.72) and 3.24 (2.67-3.83), respectively, compared with BMI 25-<30 kg/m2 (both ps for linear and quadratic terms <0.001). CONCLUSIONS Patients with MASLD and very severe obesity (BMI ≥50 kg/m2) had the highest risk, exceeding that of patients with lean MASLD, for developing liver decompensation, obesity-associated extrahepatic cancers, or dying from any cause.
Collapse
Affiliation(s)
- Jaideep Behari
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Epidemiology and Prevention Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Renwei Wang
- Cancer Epidemiology and Prevention Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Hung N. Luu
- Cancer Epidemiology and Prevention Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - David McKenzie
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michele Molinari
- Department of Surgery, Division of Transplant Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jian-Min Yuan
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Epidemiology and Prevention Program, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
33
|
Wu B, Zhang J, Chen Y, Chen S, Liu H. Association between non-alcoholic fatty liver disease and the risk of pulmonary nodules in patients with intestinal polyps. J Thorac Dis 2024; 16:3990-3999. [PMID: 38983169 PMCID: PMC11228712 DOI: 10.21037/jtd-24-754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Background Associations between metabolic risk factors and lung cancer remain elusive, and evidence on the linkage between non-alcoholic fatty liver disease (NAFLD) and pulmonary nodules is limited. This study sought to examine the independent association between NAFLD and the risk of pulmonary nodules. Methods Cross-sectional analyses of 1,119 patients with intestinal polyps hospitalized at the Department of Gastroenterology, Minhang District Central Hospital of Shanghai, China, were conducted. NAFLD was diagnosed based on hepatic ultrasonography or computed tomography (CT) findings of hepatic steatosis, with exclusion criteria ensuring patients had no history of significant alcohol consumption, viral infections, or hepatic autoimmune diseases. The currently accepted definition of a pulmonary nodule is a solid or sub-solid shadow ≤3 cm in diameter that appears as a solid or semi-solid pattern on a chest CT scan (our specific treatment is pulmonary nodule size: 5 mm to 3 cm). Adjusted 95% confidence intervals (CIs) and odds ratios (ORs) for NAFLD and the clinical features connected with pulmonary nodule risk were determined using a multivariable logistic regression analysis. Results Among the 979 intestinal polyp patients, the prevalence rates of NAFLD and pulmonary nodules were 25.9% and 32.8%, respectively. Patients with pulmonary nodules exhibited higher rates of NAFLD (31.5% vs. 23.3%, P=0.006) and obesity (41.4% vs. 32.5%, P=0.006) compared to those without pulmonary nodules. After removing all the possible confounding variables, the adjusted ORs for NAFLD, an older age, smoking, and obesity were 1.370 (95% CI: 1.006-1.867, P=0.04), 1.022 (95% CI: 1.010-1.033), 1.599 (95% CI: 1.033-2.475), and 1.410 (95% CI: 1.057-1.880), respectively (all P values <0.05). NAFLD showed a significant association with an increased risk of pulmonary nodules. Conclusions NAFLD was independently linked to an increased incidence of pulmonary nodules in intestinal polyp patients, which emphasizes the importance of screening and managing these conditions in lung cancer prevention.
Collapse
Affiliation(s)
- Bing Wu
- Department of Gastroenterology, Minhang District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Junpei Zhang
- Department of Gastroenterology, Minhang District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Ying Chen
- Department of Gastroenterology, Minhang District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Shiyao Chen
- Department of Gastroenterology, Minhang District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Hailing Liu
- Department of Gastroenterology, Minhang District Central Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Liang Y, Luo S, Bell S, Mo JMY, He B, Zhou Y, Bai X, Au Yeung SL. Do iron homeostasis biomarkers mediate the associations of liability to type 2 diabetes and glycemic traits in liver steatosis and cirrhosis: a two-step Mendelian randomization study. BMC Med 2024; 22:270. [PMID: 38926684 PMCID: PMC11210020 DOI: 10.1186/s12916-024-03486-w] [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: 12/02/2023] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Previous studies, including Mendelian randomization (MR), have demonstrated type 2 diabetes (T2D) and glycemic traits are associated with increased risk of metabolic dysfunction-associated steatotic liver disease (MASLD). However, few studies have explored the underlying pathway, such as the role of iron homeostasis. METHODS We used a two-step MR approach to investigate the associations of genetic liability to T2D, glycemic traits, iron biomarkers, and liver diseases. We analyzed summary statistics from various genome-wide association studies of T2D (n = 933,970), glycemic traits (n ≤ 209,605), iron biomarkers (n ≤ 246,139), MASLD (n ≤ 972,707), and related biomarkers (alanine aminotransferase (ALT) and proton density fat fraction (PDFF)). Our primary analysis was based on inverse-variance weighting, followed by several sensitivity analyses. We also conducted mediation analyses and explored the role of liver iron in post hoc analysis. RESULTS Genetic liability to T2D and elevated fasting insulin (FI) likely increased risk of liver steatosis (ORliability to T2D: 1.14 per doubling in the prevalence, 95% CI: 1.10, 1.19; ORFI: 3.31 per log pmol/l, 95% CI: 1.92, 5.72) and related biomarkers. Liability to T2D also likely increased the risk of developing liver cirrhosis. Genetically elevated ferritin, serum iron, and liver iron were associated with higher risk of liver steatosis (ORferritin: 1.25 per SD, 95% CI 1.07, 1.46; ORliver iron: 1.15 per SD, 95% CI: 1.05, 1.26) and liver cirrhosis (ORserum iron: 1.31, 95% CI: 1.06, 1.63; ORliver iron: 1.34, 95% CI: 1.07, 1.68). Ferritin partially mediated the association between FI and liver steatosis (proportion mediated: 7%, 95% CI: 2-12%). CONCLUSIONS Our study provides credible evidence on the causal role of T2D and elevated insulin in liver steatosis and cirrhosis risk and indicates ferritin may play a mediating role in this association.
Collapse
Affiliation(s)
- Ying Liang
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Shan Luo
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Steven Bell
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Jacky Man Yuen Mo
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Baoting He
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yangzhong Zhou
- Department of Rheumatology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, 100730, China
| | - Xiaoyin Bai
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shiu Lun Au Yeung
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
35
|
Fallah A, Abdolazimi H, Darabi Z, Talenezhad N, Mirzavandi F, Rahimpour S, Hosseinzadeh M. The association between score of plant-based diet and non-alcoholic fatty liver disease in adults. Clin Nutr ESPEN 2024; 61:407-412. [PMID: 38777462 DOI: 10.1016/j.clnesp.2024.04.013] [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: 04/22/2023] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Plant-based diet (PDI) as resource of antioxidants and anti-inflammatory phytochemicals, that was considered to protect against onset and development of Non-alcoholic fatty liver disease (NAFLD). AIM To investigate the association between plant-based diet and NAFLD in adults. METHODS The present case control study was conducted on 240 individuals (120 with NAFLD and 120 control) aged 20-69 years. Provided recommendations by the American College of Gastroenterology and the American Gastroenterological Association were used for NAFLD diagnosis. Dietary intake was assessed using 178-food item food frequency questionnaire (FFQ). Also, plant-based diet score was evaluated based on 18 food groups classified into animal foods, healthy and unhealthy plant foods. A multiple logistic regression model was used to examine the relationship between fatty liver disease and tertiles of PDI. RESULTS The results of this study showed that we did not observe any association between tertiles of PDI and NAFLD in crude model (OR: 1.29, 95%CI:0.66-2.52, P:0.44) and after adjustment for confounders including age, energy intake, physical activity, body mass index (OR:0.76, 95%CI: 0.31-1.86, P:0.52). Also, there were not any association of tertiles of healthy PDI (hPDI) (OR:1.14, 95%CI: 0.50-2.60, P:0.74) and unhealthy PDI (uhPDI) (OR:0.89, 95%CI:0.36-2.18, P: 0.79) with NAFLD after full adjustment for potential confounders. CONCLUSION There was not any association of PDI, hPDI, and uPDI with NAFLD in adults. More research needs to examine whether this specific diet may impact and improve NAFLD.
Collapse
Affiliation(s)
- Azadeh Fallah
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Abdolazimi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Darabi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nasir Talenezhad
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farhang Mirzavandi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shahab Rahimpour
- Gastroentrology Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Hosseinzadeh
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
36
|
Yu L, Gao F, Li Y, Su D, Han L, Li Y, Zhang X, Feng Z. Role of pattern recognition receptors in the development of MASLD and potential therapeutic applications. Biomed Pharmacother 2024; 175:116724. [PMID: 38761424 DOI: 10.1016/j.biopha.2024.116724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become one of the most prevalent liver diseases worldwide, and its occurrence is strongly associated with obesity, insulin resistance (IR), genetics, and metabolic stress. Ranging from simple fatty liver to metabolic dysfunction-associated steatohepatitis (MASH), even to severe complications such as liver fibrosis and advanced cirrhosis or hepatocellular carcinoma, the underlying mechanisms of MASLD progression are complex and involve multiple cellular mediators and related signaling pathways. Pattern recognition receptors (PRRs) from the innate immune system, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), NOD-like receptors (NLRs), RIG-like receptors (RLRs), and DNA receptors, have been demonstrated to potentially contribute to the pathogenesis for MASLD. Their signaling pathways can induce inflammation, mediate oxidative stress, and affect the gut microbiota balance, ultimately resulting in hepatic steatosis, inflammatory injury and fibrosis. Here we review the available literature regarding the involvement of PRR-associated signals in the pathogenic and clinical features of MASLD, in vitro and in animal models of MASLD. We also discuss the emerging targets from PRRs for drug developments that involved agent therapies intended to arrest or reverse disease progression, thus enabling the refinement of therapeutic targets that can accelerate drug development.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Feifei Gao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Yaoxin Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Dan Su
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Liping Han
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yueming Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Xuehan Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China; Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China.
| |
Collapse
|
37
|
Li Y, Tian X, Yu Q, Bao T, Dai C, Jiang L, Niu K, Yang J, Wang S, Wu X. Alleviation of hepatic insulin resistance and steatosis with NMN via improving endoplasmic reticulum-Mitochondria miscommunication in the liver of HFD mice. Biomed Pharmacother 2024; 175:116682. [PMID: 38703507 DOI: 10.1016/j.biopha.2024.116682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
The interaction between endoplasmic reticulum (ER) and mitochondria has been shown to play a key role in hepatic steatosis during chronic obesity. β-nicotinamide mononucleotide (NMN) has been reported to regulate obesity, however, its molecular mechanism at the subcellular level remains unclear. Here, NMN improved liver steatosis and insulin resistance in chronic high-fat diet (HFD) mice. RNA-seq showed that compared with the liver of HFD mice, NMN intervention enhanced fat digestion and absorption and stimulated the cholesterol metabolism signaling pathways, while impaired insulin resistance and the fatty acid biosynthesis signaling pathways. Mechanistically, NMN ameliorated mitochondrial dysfunction and ER oxidative stress in the liver of HFD mice by increasing hepatic nicotinamide adenine dinucleotide (NAD+) (P < 0.01) levels. This effect increased the contact sites (mitochondria-associated membranes [MAMs]) between ER and mitochondria, thereby promoting intracellular ATP (P < 0.05) production and mitigating lipid metabolic disturbances in the liver of HFD mice. Taken together, this study provided a theoretical basis for restoring metabolic dynamic equilibrium in the liver of HFD mice by increasing MAMs via the nutritional strategy of NMN supplementation.
Collapse
Affiliation(s)
- Yumeng Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Xutong Tian
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China; The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Qian Yu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Tongtong Bao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Chao Dai
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Liang Jiang
- ERA Biotechnology (Shenzhen) Co., Ltd, Shenzhen 518115, China
| | - Kaimin Niu
- Institute of Biological Resources, Jiangxi Academy of Sciences, Nanchang, China
| | - Jianying Yang
- The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Shujin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xin Wu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China; CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.
| |
Collapse
|
38
|
Ofosu-Boateng M, Shaik F, Choi S, Ekuban FA, Gebreyesus LH, Twum E, Nnamani D, Yeyeodu ST, Yadak N, Collier DM, Gyamfi MA. High-fat diet induced obesity promotes inflammation, oxidative stress, and hepatotoxicity in female FVB/N mice. Biofactors 2024; 50:572-591. [PMID: 38183321 PMCID: PMC11178471 DOI: 10.1002/biof.2028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/04/2023] [Indexed: 01/08/2024]
Abstract
Although obesity and subsequent liver injury are increasingly prevalent in women, female mouse models have generally shown resistance to high-fat diet (HFD)-induced obesity. We evaluated control and HFD-fed male and female FVB/N mice, a strain well-suited to transgenic analyses, for phenotypic, histological, and molecular markers related to control of glucose, lipids, and inflammation in serum, liver, and perigonadal white adipose tissues. Unlike many mouse models, HFD-fed FVB/N females gained more perigonadal and mesenteric fat mass and overall body weight than their male counterparts, with increased hepatic expression of lipogenic PPARγ target genes (Cd36, Fsp27, and Fsp27β), oxidative stress genes and protein (Nqo1 and CYP2E1), inflammatory gene (Mip-2), and the pro-fibrotic gene Pai-1, along with increases in malondialdehyde and serum ALT levels. Further, inherent to females (independently of HFD), hepatic antioxidant heme oxygenase-1 (HMOX1, HO-1) protein levels were reduced compared to their male counterparts. In contrast, males may have been relatively protected from HFD-induced oxidative stress and liver injury by elevated mRNA and protein levels of hepatic antioxidants BHMT and Gpx2, increased fatty acid oxidation genes in liver and adipocytes (Pparδ), despite disorganized and inflamed adipocytes. Thus, female FVB/N mice offer a valuable preclinical, genetically malleable model that recapitulates many of the features of diet-induced obesity and liver damage observed in human females.
Collapse
Affiliation(s)
- Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Fathima Shaik
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, 27707, NC
| | - Frederick A. Ekuban
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Lidya H. Gebreyesus
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Elizabeth Twum
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Daniel Nnamani
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Susan T. Yeyeodu
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, 27707, NC
- Charles River Discovery Services, Durham, NC, 27709
| | - Nour Yadak
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Daniel M. Collier
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Maxwell A. Gyamfi
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, 38163
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, 27707, NC
| |
Collapse
|
39
|
Aizenshtadt A, Wang C, Abadpour S, Menezes PD, Wilhelmsen I, Dalmao‐Fernandez A, Stokowiec J, Golovin A, Johnsen M, Combriat TMD, Røberg‐Larsen H, Gadegaard N, Scholz H, Busek M, Krauss SJK. Pump-Less, Recirculating Organ-on-Chip (rOoC) Platform to Model the Metabolic Crosstalk between Islets and Liver. Adv Healthc Mater 2024; 13:e2303785. [PMID: 38221504 PMCID: PMC11468483 DOI: 10.1002/adhm.202303785] [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: 10/31/2023] [Revised: 12/05/2023] [Indexed: 01/16/2024]
Abstract
Type 2 diabetes mellitus (T2DM), obesity, and metabolic dysfunction-associated steatotic liver disease (MASLD) are epidemiologically correlated disorders with a worldwide growing prevalence. While the mechanisms leading to the onset and development of these conditions are not fully understood, predictive tissue representations for studying the coordinated interactions between central organs that regulate energy metabolism, particularly the liver and pancreatic islets, are needed. Here, a dual pump-less recirculating organ-on-chip platform that combines human pluripotent stem cell (sc)-derived sc-liver and sc-islet organoids is presented. The platform reproduces key aspects of the metabolic cross-talk between both organs, including glucose levels and selected hormones, and supports the viability and functionality of both sc-islet and sc-liver organoids while preserving a reduced release of pro-inflammatory cytokines. In a model of metabolic disruption in response to treatment with high lipids and fructose, sc-liver organoids exhibit hallmarks of steatosis and insulin resistance, while sc-islets produce pro-inflammatory cytokines on-chip. Finally, the platform reproduces known effects of anti-diabetic drugs on-chip. Taken together, the platform provides a basis for functional studies of obesity, T2DM, and MASLD on-chip, as well as for testing potential therapeutic interventions.
Collapse
Affiliation(s)
- Aleksandra Aizenshtadt
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Chencheng Wang
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Shadab Abadpour
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
- Institute for Surgical ResearchOslo University HospitalOsloNorway
| | - Pedro Duarte Menezes
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Ingrid Wilhelmsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Andrea Dalmao‐Fernandez
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Department of PharmacyFaculty of Mathematics and Natural SciencesUniversity of OsloP.O. Box 1083Oslo0316Norway
| | - Justyna Stokowiec
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Alexey Golovin
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mads Johnsen
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Thomas M. D. Combriat
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
| | - Hanne Røberg‐Larsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Nikolaj Gadegaard
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Hanne Scholz
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mathias Busek
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Stefan J. K. Krauss
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| |
Collapse
|
40
|
Tuo L, Yan LT, Liu Y, Yang XX. Type 1 diabetes mellitus and non-alcoholic fatty liver disease: a two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1315046. [PMID: 38681765 PMCID: PMC11045944 DOI: 10.3389/fendo.2024.1315046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Background NAFLD (Nonalcoholic fatty liver disease) is becoming an increasingly common cause of chronic liver disease. Metabolic dysfunction, overweight/obesity, and diabetes are thought to be closely associated with increased NAFLD risk. However, few studies have focused on the mechanisms of NAFLD occurrence in T1DM. Methods We conducted a two-sample Mendelian randomization (MR) analysis to assess the causal association between T1DM and NAFLD with/without complications, such as coma, renal complications, ketoacidosis, neurological complications, and ophthalmic complications. Multiple Mendelian randomization methods, such as the inverse variance weighted (IVW) method, weighted median method, and MR-Egger test were performed to evaluate the causal association of T1DM and NAFLD using genome-wide association study summary data from different consortia, such as Finngen and UK biobank. Results We selected 37 SNPs strongly associated with NAFLD/LFC (at a significance level of p < 5 × 10-8) as instrumental variables from the Finnish database based on the T1DM phenotype (8,967 cases and 308,373 controls). We also selected 14/16 SNPs based on with or without complications. The results suggest that the genetic susceptibility of T1DM does not increase the risk of NAFLD (OR=1.005 [0.99, 1.02], IVW p=0.516, MR Egger p=0.344, Weighted median p=0.959, Weighted mode p=0.791), regardless of whether complications are present. A slight causal effect of T1DM without complications on LFC was observed (OR=1.025 [1.00, 1.03], MR Egger p=0.045). However, none of the causal relationships were significant in the IVW (p=0.317), Weighted median (p=0.076), and Weighted mode (p=0.163) methods. Conclusion Our study did not find conclusive evidence for a causal association between T1DM and NAFLD, although clinical observations indicate increasing abnormal transaminase prevalence and NAFLD progression in T1DM patients.
Collapse
Affiliation(s)
- Lin Tuo
- Department of Infectious Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | | | | | - Xing-xiang Yang
- Department of Infectious Disease, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
41
|
Mahmoudi SK, Tarzemani S, Aghajanzadeh T, Kasravi M, Hatami B, Zali MR, Baghaei K. Exploring the role of genetic variations in NAFLD: implications for disease pathogenesis and precision medicine approaches. Eur J Med Res 2024; 29:190. [PMID: 38504356 PMCID: PMC10953212 DOI: 10.1186/s40001-024-01708-8] [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: 06/10/2023] [Accepted: 02/01/2024] [Indexed: 03/21/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of chronic liver diseases, affecting more than one-quarter of people worldwide. Hepatic steatosis can progress to more severe forms of NAFLD, including NASH and cirrhosis. It also may develop secondary diseases such as diabetes and cardiovascular disease. Genetic and environmental factors regulate NAFLD incidence and progression, making it a complex disease. The contribution of various environmental risk factors, such as type 2 diabetes, obesity, hyperlipidemia, diet, and sedentary lifestyle, to the exacerbation of liver injury is highly understood. Nevertheless, the underlying mechanisms of genetic variations in the NAFLD occurrence or its deterioration still need to be clarified. Hence, understanding the genetic susceptibility to NAFLD is essential for controlling the course of the disease. The current review discusses genetics' role in the pathological pathways of NAFLD, including lipid and glucose metabolism, insulin resistance, cellular stresses, and immune responses. Additionally, it explains the role of the genetic components in the induction and progression of NAFLD in lean individuals. Finally, it highlights the utility of genetic knowledge in precision medicine for the early diagnosis and treatment of NAFLD patients.
Collapse
Affiliation(s)
- Seyedeh Kosar Mahmoudi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Shadi Tarzemani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Taha Aghajanzadeh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran.
| | - Mohammadreza Kasravi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran.
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, 1985714711, Iran.
| |
Collapse
|
42
|
Liu W, Zhu M, Liu J, Su S, Zeng X, Fu F, Lu Y, Rao Z, Chen Y. Comparison of the effects of monounsaturated fatty acids and polyunsaturated fatty acids on the lipotoxicity of islets. Front Endocrinol (Lausanne) 2024; 15:1368853. [PMID: 38501107 PMCID: PMC10945794 DOI: 10.3389/fendo.2024.1368853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/12/2024] [Indexed: 03/20/2024] Open
Abstract
Background Monounsaturated fatty acids (MUFAs) and polyunsaturated fatty acids (PUFAs) have been reported to combat saturated fatty acid (SFA)-induced cellular damage, however, their clinical effects on patients with metabolic diseases such as diabetes and hyperlipidemia are still controversial. Since comparative studies of the effects of these two types of unsaturated fatty acids (UFAs) are still limited. In this study, we aimed to compare the protective effects of various UFAs on pancreatic islets under the stress of SFA-induced metabolic disorder and lipotoxicity. Methods Rat insulinoma cell line INS-1E were treated with palmitic acid (PA) with or without UFAs including eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), arachidonic acid (AA), and oleic acid (OA) to determine cell viability, apoptosis, endoplasmic reticulum (ER) stress, and inflammatory. In vivo, male C57BL/6 mice were fed a 60% high-fat diet (HFD) for 12 w. Then the lard in HFD was partially replaced with fish oil (FO) and olive oil (OO) at low or high proportions of energy (5% or 20%) to observe the ameliorative effects of the UFA supplement. Results All UFAs significantly improved PA-induced cell viability impairment in INS-1E cells, and their alleviation on PA induced apoptosis, ER stress and inflammation were confirmed. Particularly, OA had better effects than EPA, DHA, and AA on attenuating cellular ER stress. In vivo, the diets with a low proportion of UFAs (5% of energy) had limited effects on HFD induced metabolic disorder, except for a slight improved intraperitoneal glucose tolerance in obese mice. However, when fed diets containing a high proportion of UFAs (20% of energy), both the FO and OO groups exhibited substantially improved glucose and lipid metabolism, such as decrease in total cholesterol (TC), low-density lipoprotein (LDL), fasting blood glucose (FBG), and fasting blood insulin (FBI)) and improvement of insulin sensitivity evidenced by intraperitoneal glucose tolerance test (IPGTT) and intraperitoneal insulin tolerance test (IPITT). Unexpectedly, FO resulted in abnormal elevation of the liver function index aspartate aminotransferase (AST) in serum. Pathologically, OO attenuated HFD-induced compensatory hyperplasia of pancreatic islets, while this effect was not obvious in the FO group. Conclusions Both MUFAs and PUFAs can effectively protect islet β cells from SFA-induced cellular lipotoxicity. In particular, both OA in vitro and OO in vivo showed superior activities on protecting islets function and enhance insulin sensitivity, suggesting that MUFAs might have greater potential for nutritional intervention on diabetes.
Collapse
Affiliation(s)
- Wen Liu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Min Zhu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyi Liu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Su
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Zeng
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Fudong Fu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyong Rao
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Department of Clinical Nutrition and Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Xie J, Gao H, Liu C, Pan Y, Xu C, Xu L. Causal association of nonalcoholic fatty liver disease with 22 extrahepatic cancers: A Mendelian randomization study. Hepatol Res 2024; 54:261-271. [PMID: 37877524 DOI: 10.1111/hepr.13980] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 10/26/2023]
Abstract
AIM It is unclear whether nonalcoholic fatty liver disease (NAFLD) acts as a direct contributing factor to multiple extrahepatic cancers. We aimed to systematically investigate the causal relationships of NAFLD with extrahepatic cancers. METHODS We conducted a two-sample Mendelian randomization analysis to assess the causal effects of NAFLD on 22 extrahepatic cancers. We examined the association of NAFLD with extrahepatic cancers using multiple methods in the largest genome-wide association study meta-analysis to date. We also replicated the analyses and performed two independent sensitivity analysis in the largest genome-wide association study of UK Biobank. RESULTS Using the weighted median method, genetically predicted NAFLD was significantly associated with female breast cancer risk (odds ratio [OR] 15.99; 95% confidence interval [CI] 9.58-26.69). Genetically predicted NAFLD is associated with cervical and laryngeal cancers using the inverse variance weighting method, and the ORs were 2.44 (95% CI 1.43-4.14) and 1.94 (95% CI 1.35-2.78), respectively. We observed that patatin-like phospholipase domain-containing protein 3-driven and transmembrane 6 superfamily member 2-driven NAFLD were associated with increased risks of leukemia, lung cancer, and prostate cancers (all with p < 0.05). Furthermore, we confirmed the causal association between NAFLD and breast cancer using five known single-nucleotide polymorphisms of NAFLD and six genome-wide association study-identified variants. The ORs of the weighted median estimator was 10.76 (95% CI 8.27-13.98) and 10.76 (95% CI 8.25-14.04), respectively (p < 0.001). CONCLUSION Genetically predicted NAFLD is associated with an increased risk of female breast cancer, as well as cervical, laryngeal, leukemia, lung, and prostate cancers.
Collapse
Affiliation(s)
- Jiarong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Gao
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Cenqin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yue Pan
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Gastroenterology, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
| | - Chengfu Xu
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Gastroenterology, Zhejiang Provincial Clinical Research Center for Digestive Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
44
|
Man S, Deng Y, Ma Y, Yang X, Wang X, Fu J, Yu C, Lv J, Du J, Wang B, Li L. Association between weight change, waist circumference change, and the risk of nonalcoholic fatty liver disease in individuals with metabolically healthy overweight or obesity and metabolically unhealthy overweight or obesity. Obes Res Clin Pract 2024; 18:109-117. [PMID: 38443283 DOI: 10.1016/j.orcp.2024.02.007] [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: 09/28/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND This study aimed to explore and compare the effect of weight change, and waist circumference (WC) change, on the risk of nonalcoholic fatty liver disease (NAFLD) in individuals with metabolically healthy overweight or obesity (MHOW/O) and metabolically unhealthy overweight or obesity (MUOW/O) in a health check-up cohort in China. METHODS 5625 adults with overweight or obesity, and free from NAFLD at baseline were included. Metabolically healthy was defined as not having any components of metabolic syndrome. Weight/WC changes were calculated as the relative difference between the first and second visits of check-up. NAFLD was assessed based on abdominal ultrasound. RESULTS During a median follow-up of 2.1 (IQR: 1.1-4.3) years, 1849 participants developed NAFLD. In MHOW/O participants, the multivariable adjusted HRs (95 % CIs) for NAFLD in weight change ≤ -5.0 %, and - 4.9-- 1.0 % were 0.36 (0.23-0.59), 0.59 (0.43-0.80), respectively, compared to the weight stable group (-0.9% to 0.9 %). The corresponding HRs (95 % CIs) for the association between WC change (≤ 6.0 %, - 5.9 to -3.0 %) and NAFLD in MHOW/O participants were 0.41 (0.27-0.62), and 0.74 (0.54-1.01), respectively, compared to the WC stable group (-2.9-2.9 %). Similar patterns were observed in MUOW/O participants. A more marked gradient of cumulative incidence of NAFLD across weight/WC change categories was observed in MHOW/O than in MUOW/O individuals. CONCLUSIONS A more evident association between weight/WC loss and risk of NAFLD was observed in MHOW/O than in MUOW/O individuals. Our findings indicate the practical significance of encouraging all individuals with overweight and obesity to achieve a clinically relevant level of weight/WC loss to prevent NAFLD, even among metabolic healthy groups.
Collapse
Affiliation(s)
- Sailimai Man
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China; Meinian Institute of Health, Beijing 100083, China; Peking University Health Science Center Meinian Public Health Institute, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
| | - Yuhan Deng
- Meinian Institute of Health, Beijing 100083, China; Chongqing Research Institute of Big Data, Peking University, Chongqing 400000, China
| | - Yuan Ma
- Meinian Institute of Health, Beijing 100083, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaochen Yang
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing 100191, China
| | - Xiaona Wang
- Beijing MJ Health Check-up Center, Beijing 100000, China
| | - Jingzhu Fu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China; Peking University Health Science Center Meinian Public Health Institute, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China; Peking University Health Science Center Meinian Public Health Institute, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China; Peking University Health Science Center Meinian Public Health Institute, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China
| | - Jing Du
- Beijing Centre for Disease Prevention and Control, Beijing 100013, China.
| | - Bo Wang
- Meinian Institute of Health, Beijing 100083, China; Peking University Health Science Center Meinian Public Health Institute, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China.
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China; Peking University Health Science Center Meinian Public Health Institute, Beijing 100191, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China; Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing 100191, China.
| |
Collapse
|
45
|
Otero Sanchez L, Chen Y, Lassailly G, Qi X. Exploring the links between types 2 diabetes and liver-related complications: A comprehensive review. United European Gastroenterol J 2024; 12:240-251. [PMID: 38103189 PMCID: PMC10954434 DOI: 10.1002/ueg2.12508] [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: 07/27/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
In recent decades, the prevalence of type 2 diabetes has been steadily increasing, presenting a significant global public health challenge. These epidemiological trends can be attributed to significant lifestyle changes in modern societies, characterized by sedentary behavior and the consumption of hypercaloric, highly processed foods, along with the aging of the human population. As a result, it has become crucial for both public healthcare systems and healthcare providers to prioritize the management of diabetes and identify its systemic consequences. Emerging research has shed light on the links and risks between diabetes and liver events. This comprehensive review aims to explore the complex interplay between type 2 diabetes mellitus and liver-related outcomes, especially hepatocellular carcinoma and cirrhosis, offering insights into effective methods for detecting liver risk in individuals with diabetes. Additionally, the review will assess the various treatments that could hold the potential for positive outcomes in managing both diabetes and metabolic dysfunction-associated steatotic liver disease and liver fibrosis.
Collapse
Affiliation(s)
- Lukas Otero Sanchez
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, CUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Yuping Chen
- Center of Portal Hypertension, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Guillaume Lassailly
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, CUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Service des maladies de l'appareil digestif, hôpital Huriez, CHU de Lille, Université de Lille, Lille, France
| | - Xiaolong Qi
- Center of Portal Hypertension, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
46
|
Pustake MV, Giri P, Ganiyani MAM, Jain S. Diabetes, nonalcoholic fatty liver disease and obesity: What is the link? J Family Med Prim Care 2024; 13:1119-1120. [PMID: 38736789 PMCID: PMC11086803 DOI: 10.4103/jfmpc.jfmpc_445_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/07/2023] [Indexed: 05/14/2024] Open
Affiliation(s)
- Manas V. Pustake
- Department of Internal Medicine, Grant Government Medical College and Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
- Harvard Medical School, Harvard, University, Boston, Massachusetts, USA
| | - Purushottam Giri
- Department of Community Medicine, IIMSR Medical College, Badnapur, Jalna, Maharashtra, India
| | - Mohammad Arfat M.T. Ganiyani
- Department of Internal Medicine, Grant Government Medical College and Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
- Harvard Medical School, Harvard, University, Boston, Massachusetts, USA
| | - Suhani Jain
- Department of Internal Medicine, Grant Government Medical College and Sir J. J. Group of Hospitals, Mumbai, Maharashtra, India
| |
Collapse
|
47
|
Kong L, Ye C, Wang Y, Dou C, Zheng J, Wang S, Lin H, Zhao Z, Li M, Xu Y, Chen Y, Lu J, Xu M, Wang W, Ning G, Bi Y, Wang T. Diabesity phenotype in relation to the incidence and resolution of nonalcoholic fatty liver disease: A prospective cohort study. J Diabetes 2024; 16:e13459. [PMID: 37584361 PMCID: PMC10809295 DOI: 10.1111/1753-0407.13459] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/06/2023] [Accepted: 07/30/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Diabesity is a term used to emphasize the dual epidemic and the combined detrimental effects of diabetes and obesity. We aimed to investigate the associations of diabesity with the incidence and resolution of nonalcoholic fatty liver disease (NAFLD). METHODS This prospective cohort study included 5549 participants with a median follow-up of 4.3 years (2010-2015). Diabesity was defined as six categories by the combinations of glucose tolerance status (normal glucose tolerance [NGT], prediabetes, and diabetes) diagnosed by fasting and oral glucose tolerance test 2-h glucose and hemoglobin A1c and general or abdominal obesity status. We examined the odds ratios (ORs) for the incidence and resolution of NAFLD associated with diabesity categories, respectively. RESULTS For NAFLD incidence, compared with the diabesity category of NGT with nonobesity, the categories of either glucose intolerance or general obesity were associated with higher risks of NAFLD, of which the categories with obesity, regardless of glucose intolerance status, exhibited greater risks (ORs ranged from 3.19 to 4.49) than the categories of nonobesity. For NAFLD resolution, the categories of prediabetes or diabetes with obesity were associated with decreased likelihoods of a resolution of NAFLD (ORs ranged from 0.40 to 0.58). These association patterns were consistent across various definitions of diabesity by glucose tolerance status diagnosed by different combinations of glycemic parameters and general or abdominal obesity. CONCLUSIONS The diabesity association pattern with NAFLD incidence was mainly determined by obesity, while that with NAFLD resolution was driven by the combined phenotype of glucose intolerance and obesity.
Collapse
Affiliation(s)
- Lijie Kong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chaojie Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yiying Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chun Dou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
48
|
Liu W, Hu C, Zhang J, Qian X, Jiang Y, Liang K, An R, Pan L, Wang X. Simultaneous determination of fatty acids and related N-Acylethanolamines in rat plasma, rat tissue, and human plasma using UPLC-MS/MS. Microchem J 2024; 196:109633. [DOI: 10.1016/j.microc.2023.109633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
|
49
|
Deng Y, Huang J, Wong MCS. Associations of non-alcoholic fatty liver disease and cirrhosis with liver cancer in European and East Asian populations: A Mendelian randomization study. Cancer Rep (Hoboken) 2024; 7:e1913. [PMID: 37840448 PMCID: PMC10809194 DOI: 10.1002/cnr2.1913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/27/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND The positive relationships of non-alcoholic fatty liver disease (NAFLD) and cirrhosis with liver cancer were shown in previous observational studies, while further Mendelian randomization (MR) investigations are needed to confirm the possible causal associations. AIMS This study aimed to explore whether NAFLD and cirrhosis were causally related to liver cancer using MR in European and East Asian populations. METHODS AND RESULTS For European populations, NAFLD data were obtained from a genome-wide meta-analysis (8434 patients and 770 180 controls). The data on chronic elevation of alanine aminotransferase (cALT), a proxy of NAFLD, were derived from Million Veteran Program (68 725 patients and 95 472 controls). Cirrhosis data were collected from two sources: a genome-wide association study of five cohorts (4829 patients and 72 705 controls) and FinnGen (1931 patients and 216 861 controls). Liver cancer data were collected from FinnGen (304 patients and 174 006 controls). For East Asian populations, the data on cirrhosis (2184 patients and 210 269 controls) and hepatocellular carcinoma (1866 patients and 195 745 controls) were obtained from Biobank Japan. Three, 41, seven, six, and three single-nucleotide polymorphisms were used for NAFLD (European), cALT (European), cirrhosis (European-five cohorts), cirrhosis (European-FinnGen), and cirrhosis (East Asian), respectively. We used inverse-variance weighted as the primary method to calculate the odds ratio (OR) and 95% confidence interval (CI). Among European populations, genetically-predicted NAFLD, cALT, cirrhosis (five cohorts), and cirrhosis (FinnGen) were positively associated with liver cancer, with ORs (95% CIs) of 6.62 (3.81-11.50) (p < .001), 2.59 (1.70-3.94) (p < .001), 3.38 (2.41-4.75) (p < .001), and 2.62 (1.20-5.72) (p = .015). Among East Asian populations, there was also a positive association between genetically-predicted cirrhosis and hepatocellular carcinoma (OR = 2.12; 95% CI = 1.78-2.52; p < .001). CONCLUSION This study utilized MR to complementarily confirm the positive connections of NAFLD and cirrhosis with liver cancer, as identified in earlier observational research. Subsequent MR investigations involving more liver cancer cases are needed.
Collapse
Affiliation(s)
- Yunyang Deng
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
| | - Martin Chi Sang Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of MedicineThe Chinese University of Hong KongHong Kong SARChina
- School of Public HealthThe Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- School of Public HealthPeking UniversityBeijingChina
| |
Collapse
|
50
|
Shakour N, Karami S, Iranshahi M, Butler AE, Sahebkar A. Antifibrotic effects of sodium-glucose cotransporter-2 inhibitors: A comprehensive review. Diabetes Metab Syndr 2024; 18:102934. [PMID: 38154403 DOI: 10.1016/j.dsx.2023.102934] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/25/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND AND AIMS Scar tissue accumulation in organs is the underlying cause of many fibrotic diseases. Due to the extensive array of organs affected, the long-term nature of fibrotic processes and the large number of people who suffer from the negative impact of these diseases, they constitute a serious health problem for modern medicine and a huge economic burden on society. Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a relatively new class of anti-diabetic pharmaceuticals that offer additional benefits over and above their glucose-lowering properties; these medications modulate a variety of diseases, including fibrosis. Herein, we have collated and analyzed all available research on SGLT2is and their effects on organ fibrosis, together with providing a proposed explanation as to the underlying mechanisms. METHODS PubMed, ScienceDirect, Google Scholar and Scopus were searched spanning the period from 2012 until April 2023 to find relevant articles describing the antifibrotic effects of SGLT2is. RESULTS The majority of reports have shown that SGLT2is are protective against lung, liver, heart and kidney fibrosis as well as arterial stiffness. According to the results of clinical trials and animal studies, many SGLT2 inhibitors are promising candidates for the treatment of fibrosis. Recent studies have demonstrated that SGLT2is affect an array of cellular processes, including hypoxia, inflammation, oxidative stress, the renin-angiotensin system and metabolic activities, all of which have been linked to fibrosis. CONCLUSION Extensive evidence indicates that SGLT2is are promising treatments for fibrosis, demonstrating protective effects in various organs and influencing key cellular processes linked to fibrosis.
Collapse
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Karami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|