1
|
Carbone F, Després JP, Ioannidis JPA, Neeland IJ, Garruti G, Busetto L, Liberale L, Ministrini S, Vilahur G, Schindler TH, Macedo MP, Di Ciaula A, Krawczyk M, Geier A, Baffy G, Faienza MF, Farella I, Santoro N, Frühbeck G, Yárnoz-Esquiroz P, Gómez-Ambrosi J, Chávez-Manzanera E, Vázquez-Velázquez V, Oppert JM, Kiortsis DN, Sbraccia P, Zoccali C, Portincasa P, Montecucco F. Bridging the gap in obesity research: A consensus statement from the European Society for Clinical Investigation. Eur J Clin Invest 2025:e70059. [PMID: 40371883 DOI: 10.1111/eci.70059] [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: 01/09/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Most forms of obesity are associated with chronic diseases that remain a global public health challenge. AIMS Despite significant advancements in understanding its pathophysiology, effective management of obesity is hindered by the persistence of knowledge gaps in epidemiology, phenotypic heterogeneity and policy implementation. MATERIALS AND METHODS This consensus statement by the European Society for Clinical Investigation identifies eight critical areas requiring urgent attention. Key gaps include insufficient long-term data on obesity trends, the inadequacy of body mass index (BMI) as a sole diagnostic measure, and insufficient recognition of phenotypic diversity in obesity-related cardiometabolic risks. Moreover, the socio-economic drivers of obesity and its transition across phenotypes remain poorly understood. RESULTS The syndemic nature of obesity, exacerbated by globalization and environmental changes, necessitates a holistic approach integrating global frameworks and community-level interventions. This statement advocates for leveraging emerging technologies, such as artificial intelligence, to refine predictive models and address phenotypic variability. It underscores the importance of collaborative efforts among scientists, policymakers, and stakeholders to create tailored interventions and enduring policies. DISCUSSION The consensus highlights the need for harmonizing anthropometric and biochemical markers, fostering inclusive public health narratives and combating stigma associated with obesity. By addressing these gaps, this initiative aims to advance research, improve prevention strategies and optimize care delivery for people living with obesity. CONCLUSION This collaborative effort marks a decisive step towards mitigating the obesity epidemic and its profound impact on global health systems. Ultimately, obesity should be considered as being largely the consequence of a socio-economic model not compatible with optimal human health.
Collapse
Affiliation(s)
- Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Jean-Pierre Després
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, Québec, Canada
- VITAM - Centre de Recherche en santé Durable, Centre intégré Universitaire de santé et de Services Sociaux de la Capitale-Nationale, Québec, Québec, Canada
| | - John P A Ioannidis
- Department of Medicine, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Biomedical Science, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
| | - Ian J Neeland
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Cardiovascular Disease, Harrington Heart and Vascular Institute, Cleveland, Ohio, USA
| | - Gabriella Garruti
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Luca Busetto
- Department of Medicine, University of Padua, Padua, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Cardiology Department, Luzerner Kantonspital, Lucerne, Switzerland
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Thomas H Schindler
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Paula Macedo
- APDP - Diabetes Portugal, Education and Research Center, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS | FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Agostino Di Ciaula
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Andreas Geier
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital of Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Hepatology, University Hospital of Würzburg, Würzburg, Germany
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Yárnoz-Esquiroz
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Chávez-Manzanera
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jean-Michel Oppert
- Department of Nutrition, Pitié-Salpêtrière Hospital (AP-HP), Human Nutrition Research Center Ile-de-France (CRNH IdF), Sorbonne University, Paris, France
| | - Dimitrios N Kiortsis
- Atherothrombosis Research Centre, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Piero Portincasa
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| |
Collapse
|
2
|
Xu M, Gong R, Xie J, Xu S, Wang S. Clinical characteristics of lean and non-lean non-alcoholic fatty liver disease: a cross-sectional study. Nutr Metab (Lond) 2025; 22:40. [PMID: 40355898 PMCID: PMC12070601 DOI: 10.1186/s12986-025-00927-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) affects more than a quarter of the global population and has become the world's number one chronic liver disease, seriously jeopardizing public life and health. Despite the new terminology of metabolic dysfunction-associated steatotic liver disease (MASLD) has been proposed, the mechanisms underlying the heterogeneity across BMI stratification in non-alcoholic fatty liver disease (NAFLD) remain unclear. The aim of this study was to reveal the differences in metabolic and fibrotic characteristics between lean (BMI < 23 kg/m2) and non-lean NAFLD in an Asian population. METHODS The current study collected NAFLD patients from the physical examination population. Patients were divided into two groups by BMI to compare their clinical parameters, including lean (BMI < 23 kg/m2) and non-lean (BMI ≥ 23 kg/m2) and fibrosis subgroups (with a threshold of LSM = 8 kPa) and analyzed for risk factors by logistic regression models. RESULTS Of the 11,577 NAFLD patients who participated in the study, there were 916 lean and 10,661 non-lean. The non-lean group was younger than the lean group (median age 50 vs. 52 years, P < 0.001) and had a significantly higher prevalence of hypertension (28.0% vs. 18.3%), diabetes mellitus (10.1% vs. 6.1%), and liver fibrosis (9.1% vs. 5.1%) (all P < 0.001). Analysis of metabolic indexes showed that TyG, TyG-BMI, TG/HDL-C and APRI were higher in the non-lean group (all P < 0.001). Gender stratification revealed that ALT was significantly higher in the male non-lean group, while HDL-C was lower in the female non-lean group (1.35 vs. 1.47 mmol/L). Multiple regression suggested that the risk of fibrosis was independently associated with CAP values and fasting glucose, BMI, direct bilirubin, globulin, and age in the non-lean group, whereas the risk was mainly driven by GGT and ALP in the lean group. CONCLUSIONS Non-lean NAFLD patients showed more significant metabolic disturbances and risk of liver fibrosis. Although metabolic indicators (TyG, FIB-4) have limited predictive value for liver fibrosis, they are strongly associated with metabolic risk in MASLD.
Collapse
Affiliation(s)
- Mengyan Xu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rui Gong
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiao Xie
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sanping Xu
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Shi Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Lu L, Chen Y, Liu B, Li X, Wang J, Nie Z, Fu X. Association between cumulative changes of the triglyceride glucose index and incidence of stroke in a population with cardiovascular-kidney-metabolic syndrome stage 0-3: a nationwide prospective cohort study. Cardiovasc Diabetol 2025; 24:202. [PMID: 40355933 PMCID: PMC12070779 DOI: 10.1186/s12933-025-02754-0] [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: 03/12/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The triglyceride-glucose (TyG) index was associated with higher risk of mortality in individuals with Cardiovascular-Kidney-Metabolic (CKM) syndrome stages 0-3. However, the relationship between cumulative of TyG (cumTyG) and incidence of stroke remains unclear in individuals with CKM syndrome stages 0-3. METHOD Participants with CKM syndrome stage 0-3 were enrolled from the China Health and Retirement Longitudinal Study (CHARLS) from 2011 to 2015. TyG was calculated as ln [fasting triglyceride (mg/dL)×fasting glucose (mg/dL)/2], and the cumTyG, as an area-under-the-curve estimate (mean TyG × time span), was calculated as (TyG2012 + TyG 2015)/2 * time (2015-2012). TyG control levels were classified using k-mean clustering analysis. Logistic regression was used to analyze the effect of cumTyG and TyG control levels on the incidence of stroke. Restricted cubic spline models (RCS) were performed to explore the potential non-linear relationship between cumTyG and stroke risk at different CKM syndrome stages 0-3. RESULTS A total of 4,700 CKM syndrome stages 0-3 participants were enrolled, among 280 patients had developed stroke during the 3-year follow-up period. After adjusting for confounders, compared to class 1 group, the odds ratio (OR) of incidents of stroke for class 2 was 1.39 [95% confidence interval (CI) 1.003, 1.92], P = 0.046; the OR of incidents of stroke for class 3 was 1.28 (95% CI 0.92-1.77), P = 0.147, the OR of incidents of stroke for class 4 was 1.28 (95% CI 0.84-1.94), P = 0.238. Elevated cumTyG was associated with an increase in incidence of stroke (OR 1.13, 95% CI 1.05, 1.22, P = 0.002). The relationship between the cumTyG index and stroke was linear in restricted cubic spline regression. CONCLUSIONS Elevated cumTyG was associated with an increased risk of stroke events in the population of CKM syndrome stages 0-3. Long-term dynamic monitoring of changes of TyG may help in the early identification of patients at high risk of developing stroke in the individuals with CKM syndrome stages 0-3.
Collapse
Affiliation(s)
- Lifei Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Yubiao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
- Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Baiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
- Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Xicong Li
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan, People's Republic of China
- Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Jiale Wang
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, 1st Xinzao Rd, Xinzao, Panyu District, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Zhengchang Nie
- 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan, People's Republic of China
- Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China
| | - Xiaodong Fu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, 1st Xinzao Rd, Xinzao, Panyu District, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Chen YW, Ahn IS, Wang SSM, Majid S, Diamante G, Cely I, Zhang G, Cabanayan A, Komzyuk S, Bonnett J, Arneson D, Yang X. Multitissue single-cell analysis reveals differential cellular and molecular sensitivity between fructose and high-fat high-sucrose diets. Cell Rep 2025; 44:115690. [PMID: 40349341 DOI: 10.1016/j.celrep.2025.115690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/03/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic syndrome (MetS), a conglomerate of dysregulated metabolic traits that vary between individuals, is partially driven by modern diets high in fat, sucrose, or fructose and their interactions with host genes in metabolic tissues. To elucidate the roles of individual tissues and cell types in diet-induced MetS, we performed single-cell RNA sequencing on the hypothalamus, liver, adipose tissue, and small intestine of mice fed high-fat high-sucrose (HFHS) or fructose diets. We found that hypothalamic neurons were sensitive to fructose, while adipose progenitor cells and macrophages were responsive to HFHS. Ligand-receptor analysis revealed lipid metabolism and inflammation networks among peripheral tissues driven by HFHS, while both diets stimulated synaptic remodeling within the hypothalamus. mt-Rnr2, a top responder to both diets, mitigated diet-induced MetS by stimulating thermogenesis. Our study demonstrates that HFHS and fructose diets have differential cell type and network targets but also share regulators such as mt-Rnr2 to affect MetS risk.
Collapse
Affiliation(s)
- Yen-Wei Chen
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Molecular Toxicology, University of California, Los Angeles, Los Angeles, CA, USA
| | - In Sook Ahn
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Susanna Sue-Ming Wang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sana Majid
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Graciel Diamante
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ingrid Cely
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Molecular Toxicology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Guanglin Zhang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Angelus Cabanayan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sergey Komzyuk
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jack Bonnett
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Douglas Arneson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Molecular Toxicology, University of California, Los Angeles, Los Angeles, CA, USA; Interdepartmental Program of Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Torpey K, Ganu V, Kenu E, Ayisi Addo S, Agyabeng K, Odikro MA, Adu-Gyamfi R, Mohammed AG, Lartey M. Metabolic syndrome among a Ghanaian cohort living with HIV initiated on dolutegravir in a real-world setting: a prospective study. BMJ Open 2025; 15:e097340. [PMID: 40345689 PMCID: PMC12067840 DOI: 10.1136/bmjopen-2024-097340] [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: 11/29/2024] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES The use of antiretroviral therapy has been linked to the development of some components of metabolic syndrome (MetS), specifically glucose intolerance, weight gain and defective lipid metabolism. This study determined the relationship between dolutegravir (DTG) and MetS in a cohort of persons living with HIV (PWH) initiating DTG-based regimen in Ghana. DESIGN A 2-year observational prospective study was conducted from September 2020 to August 2022. SETTING Five HIV high-burden facilities providing antiretroviral therapy services at the district and tertiary levels of care in Ghana. PARTICIPANTS Persons with HIV who were newly enrolled onto DTG. PRIMARY AND SECONDARY OUTCOME MEASURES Waist circumference, body mass index, blood pressure, fasting blood glucose and lipids were the primary outcomes measured at baseline, 3, 6, 12 and at 18 months follow-up to determine the incidence of MetS. MetS was defined using the Joint Consensus definition that combines the International Diabetes Federation and the National Cholesterol Education Programme Adult Treatment Panel III (ATP III) definitions. The Kaplan-Meier estimator was used to estimate the risk of developing MetS. The Cox proportional hazard model was used in estimating HRs. RESULTS Of 3664 PWH screened at baseline, 31.4% (1152/3664) had MetS. Of the remaining 2512 with no MetS at baseline, there were 960 incident cases of MetS over the 1.5 years follow-up. The estimated MetS incident rate is 384.2 (95% CI: 360.6 to 409.2) per 1000 person-years with a median time to development of MetS at 6 months (IQR; 3-12 months). Being female (adjsuted HR, aHR: 1.42, 95% CI: 1.19 to 1.70), age ≥50 years (aHR: 1.30, 95% CI: 1.12 to 1.51), having a comorbidity at baseline (aHR: 1.39, 95% CI: 1.12 to 1.51) and being overweight (aHR: 1.46, 95% CI: 1.25 to 1.71) and obese (aHR: 1.62, 95% CI: 1.36 to 1.93) were associated with higher risk of MetS development. CONCLUSIONS The incidence of MetS was high among our patients, with elevated fasting blood sugar and elevated blood pressure being the most common developed MetS defining components. HIV programmes should institute targeted interventions at addressing central obesity to reduce the risk of MetS.
Collapse
Affiliation(s)
- Kwasi Torpey
- Department of Population, Family, Reproductive Health, School of Public health, University of Ghana, Accra, Ghana
| | - Vincent Ganu
- Department of Medicine, Korle Bu Teaching Hospital, Accra, Ghana
| | - Ernest Kenu
- Ghana Field Epidemiology and Laboratory Training Programme, Department of Epidemiology and Disease Control, School of Public Health, University of Ghana, Accra, Ghana
| | - Stephen Ayisi Addo
- National AIDS &STI Control Programme, Ghana Health Service, Accra, Ghana
| | - Kofi Agyabeng
- Department of Biostatistics, School of Public Health, University of Ghana, Accra, Ghana
| | - Magdalene Akos Odikro
- Ghana Field Epidemiology and Laboratory Training Programme, Department of Epidemiology and Disease Control, School of Public Health, University of Ghana, Accra, Ghana
| | - Raphael Adu-Gyamfi
- National AIDS &STI Control Programme, Ghana Health Service, Accra, Ghana
| | | | - Margaret Lartey
- Department of Medicine, Korle Bu Teaching Hospital, Accra, Ghana
- Department of Medicine & Therapeutics, University of Ghana Medical School, Accra, Ghana
| |
Collapse
|
6
|
El-Shoukrofy MS, Ismail A, Elhamammy RH, Abdelhady SA, Nassra R, Makkar MS, Agami MA, Wahid A, Nematalla HA, Sai M, Merk D, El-Yazbi AF, Belal ASF, Eid AH, Elzahhar PA. Novel thiazolones for the simultaneous modulation of PPARγ, COX-2 and 15-LOX to address metabolic disease-associated portal inflammation. Eur J Med Chem 2025; 289:117415. [PMID: 40022874 DOI: 10.1016/j.ejmech.2025.117415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
A hybrid pharmacophore model, based on structural motifs previously identified by our team, was employed to generate ligands that simultaneously target COX-2, 15-LOX, and PPARγ in the context of metabolic dysfunction-associated fatty liver disease (MAFLD). Notable COX-2 inhibitory activities (IC50 = 0.065-0.24 μM) were observed relative to celecoxib (IC50 = 0.049 μM). The two most effective 15-LOX inhibitors, 2a and 2b, exhibited 69 % and 57 % of quercetin's action, respectively. Utilizing the rat hemi-diaphragm model to assess in vitro glucose uptake capacity, compounds 2a and 2b demonstrated significant glucose uptake potential in the absence of insulin, surpassing that of pioglitazone. Compound 2a activated PPARγ with an EC50 value of 3.4 μM in a Gal4-hybrid reporter gene assay, indicating partial agonistic action. Interesting binding interactions with targets of interest were identified by molecular docking studies. As well, the expression levels of 20-HETE, Il-1β and TNF-α were decreased in LPS-challenged RAW264.7 macrophages upon treatment with compound 2a. The pharmacokinetic analysis of 2a and assessment of its in vivo efficacy in addressing hepatic impairment in rat models of diabetes and pre-diabetes were carried out. Together, these findings may offer preliminary insights into the potential of these compounds for further refinement in the existing therapeutic arsenals for metabolic diseases.
Collapse
Affiliation(s)
- Mai S El-Shoukrofy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Azza Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Reem H Elhamammy
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Sherien A Abdelhady
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza Complex 21648, Alexandria, Egypt
| | - Rasha Nassra
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, 21131, Egypt
| | - Monica S Makkar
- Faculty of Pharmacy and the Research & Innovation Hub, Alamein International University, Alamein, 5060335, Egypt
| | - Mahmoud A Agami
- Faculty of Pharmacy and the Research & Innovation Hub, Alamein International University, Alamein, 5060335, Egypt
| | - Ahmed Wahid
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, 22516, Egypt
| | - Minh Sai
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377, Munich, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438, Frankfurt, Germany
| | - Ahmed F El-Yazbi
- Faculty of Pharmacy and the Research & Innovation Hub, Alamein International University, Alamein, 5060335, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed S F Belal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, 2713, Qatar.
| | - Perihan A Elzahhar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| |
Collapse
|
7
|
Pak YK, Im S, Choi HS, Lind L, Lind M, Lee HK. Correlation between environmental pollutant exposure and cardiopulmonary health by serum biomarker analysis in the Swedish elderly population. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025; 35:1156-1169. [PMID: 39037202 DOI: 10.1080/09603123.2024.2382306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Persistent organic pollutants (POPs) affect human health through the aryl hydrocarbon receptor (AhR) pathway and are implicated in mitochondrial dysfunction. Using data from the PIVUS study, we investigated the associations of serum AhR ligand (POP)-mediated luciferase activity (AhRL), mitochondrial ATP production inhibiting substances (MIS-ATP), and those affecting reactive oxygen species (MIS-ROS) with several metabolic syndrome (MetS) and cardiopulmonary function parameters. These include insulin resistance (HOMA-IR), inflammation, oxidative stress, and cardiopulmonary variables (FVC, FEV1, LV-EF, CCA distensibility). MIS-ATP showed significant correlations with HOMA-IR and pulmonary functions, indicating its direct impact of MIS-ATP on metabolic and pulmonary health. MIS-ROS correlated with oxidative stress markers and CCA distensibility, suggesting a role in systemic inflammatory responses. This study highlights the intricate relationships between environmental pollutant mixture and cardiopulmonary health in MetS as indicated by biomarkers of POP exposure in the elderly population, suggesting POP exposure may influence MetS onset and progression through mitochondrial dysfunction.
Collapse
Affiliation(s)
- Youngmi Kim Pak
- Department of Physiology, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul, South Korea
- Department of Neuroscience, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Suyeol Im
- Department of Neuroscience, Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Hoon Sung Choi
- Department of Internal Medicine, Chung Ang University College of Medicine, Seoul, South Korea
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Monica Lind
- Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Hong Kyu Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
8
|
Byrne CD, Armandi A, Pellegrinelli V, Vidal-Puig A, Bugianesi E. Μetabolic dysfunction-associated steatotic liver disease: a condition of heterogeneous metabolic risk factors, mechanisms and comorbidities requiring holistic treatment. Nat Rev Gastroenterol Hepatol 2025; 22:314-328. [PMID: 39962331 DOI: 10.1038/s41575-025-01045-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/09/2025]
Abstract
Μetabolic dysfunction-associated steatotic liver disease (MASLD) comprises a heterogeneous condition in the presence of steatotic liver. There can be a hierarchy of metabolic risk factors contributing to the severity of metabolic dysfunction and, thereby, the associated risk of both liver and extrahepatic outcomes, but the precise ranking and combination of metabolic syndrome (MetS) traits that convey the highest risk of major adverse liver outcomes and extrahepatic disease complications remains uncertain. Insulin resistance, low-grade inflammation, atherogenic dyslipidaemia and hypertension are key to the mechanisms of liver and extrahepatic complications. The liver is pivotal in MetS progression as it regulates lipoprotein metabolism and secretes substances that affect insulin sensitivity and inflammation. MASLD affects the kidneys, heart and the vascular system, contributing to hypertension and oxidative stress. To address the global health burden of MASLD, intensified by obesity and type 2 diabetes mellitus epidemics, a holistic, multidisciplinary approach is essential. This approach should focus on both liver disease management and cardiometabolic risk factors. This Review examines the link between metabolic dysfunction and liver dysfunction and extrahepatic disease outcomes, the diverse mechanisms in MASLD due to metabolic dysfunction, and a comprehensive, personalized management model for patients with MASLD.
Collapse
Affiliation(s)
- Christopher D Byrne
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton, UK
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vanessa Pellegrinelli
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, MRC MDU Unit, University of Cambridge, Cambridge, UK
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
9
|
Sun J, Shi R, Zhou Z, Xu W, Huai J, Cao Y, Zhang W, Nie L, Wang G, Yan Q, Wang X, Li M, Fang Z, Zhou X. Identification of CACNB1 protein as an actionable therapeutic target for hepatocellular carcinoma via metabolic dysfunction analysis in liver diseases: An integrated bioinformatics and machine learning approach for precise therapy. Int J Biol Macromol 2025; 308:142315. [PMID: 40139615 DOI: 10.1016/j.ijbiomac.2025.142315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
In addition to histological evaluation for nonalcoholic fatty liver disease (NAFLD), a comprehensive analysis of the metabolic landscape is urgently needed to categorize patients into distinct subgroups for precise treatment. In this study, a total of 806 NAFLD and 267 normal liver samples were comprehensively analyzed. Alterations in 114 metabolic pathways were investigated and two distinct metabolic clusters were identified. Single-cell RNA sequencing (scRNA-seq) analysis was utilized to decipher the metabolic activities within the microenvironment of NAFLD-derived liver cirrhosis. A refined fibrosis prediction model was developed using a Gaussian Mixture Model (GMM), demonstrating superior performance in fibrosis discrimination across multiple independent cohorts. Additionally, using The Cancer Genome Atlas (TCGA), CACNB1 protein was identified as a promising therapeutic target for hepatocellular carcinoma (HCC) patients with elevated metabolic dysfunction scores (MBDS). Machine learning algorithms were applied to MBDS-related genes to select an optimal prognostic model for HCC. All the models were trained in an HCC cohort obtained from the Gene Expression Omnibus (GEO), and the best model was validated in two independent HCC datasets: the TCGA-HCC cohort and LIRI-JP cohort. Overall, we provide insights of metabolic molecular subtyping and its potential clinical applicability in risk stratification for NAFLD and HCC individuals.
Collapse
Affiliation(s)
- Jing Sun
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaokai Zhou
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Weilong Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaxuan Huai
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yutian Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenhui Zhang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lijuan Nie
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Gaoxiang Wang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qianhua Yan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacology, Renmin Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Minglun Li
- Department of Radiation Oncology, Lueneburg Municipal Hospital, Lueneburg, Germany.
| | - Zhuyuan Fang
- Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xiqiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
10
|
Eslami Z, Joshaghani H, Eghbal Moghanlou A, Norouzi A, Mirghani SJ. Atorvastatin and flaxseed dietary treatments improve dyslipidemia and liver injuries in a diet-induced rat model of non-alcoholic fatty liver disease. AVICENNA JOURNAL OF PHYTOMEDICINE 2025; 15:1102-1112. [PMID: 40365191 PMCID: PMC12068500 DOI: 10.22038/ajp.2024.25220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/17/2024] [Indexed: 05/15/2025]
Abstract
Objective Non-alcoholic fatty liver disease (NAFLD) as the most common chronic liver disease is associated with metabolic disorders including dysregulated lipid and glucose metabolism. There is no approved drug treatment for NAFLD; thus, new therapies are needed. We studied the antidyslipidemic effects of atorvastatin and/or possibly hepatoprotective effects of flaxseed/ flaxseed oil in a rat model of NAFLD. Materials and Methods Fifty-six male Wistar rats were divided randomly into seven groups: 1) control, 2) high-fructose diet (HFD), 3) HFD +atorvastatin (20 mg/kg), 4) HFD+ flaxseed (40 g/kg), 5) HFD+ flaxseed oil (40 mg/kg), 6) HFD+flaxseed (40 g/kg) + atorvastatin (20 mg/kg) and 7) HFD+flaxseed oil (40 g/kg) +atorvastatin (20 mg/kg). The interventions were done for 23 weeks, after which anthropometric indices, lipid profile, liver enzymes, fasting blood glucose, and kidney indices were analyzed. Scoring of hematoxylin-eosin-stained liver sections was used to assess the severity of NAFLD. Results All the treatments reduced mesenteric fat mass, and the amount of fat around the liver, except in HFD+ flaxseed +atorvastatin group. The interventions improved NAFLD activity score, which considers steatosis, lobular inflammation, and hepatocyte ballooning. However, atorvastatin was most efficient in reducing inflammation and hepatocyte ballooning. While atorvastatin reduced only Gamma-glutamyltransferase (GGT) levels, flaxseed or flaxseed oil mono- and combination therapies reduced serum levels of all liver enzymes. The interventions improved the serum lipid profile and all, except atorvastatin decreased fasting blood glucose. Conclusion Flaxseed therapies improved NAFLD-associated liver injuries and dyslipidemia, while atorvastatin mostly reduced hepatocyte ballooning and lobular inflammation.
Collapse
Affiliation(s)
- Zahra Eslami
- Department of Clinical Biochemistry, Hamadan University of medical science, Hamadan, Iran
| | - Hamidreza Joshaghani
- Laboratory sciences research center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdorreza Eghbal Moghanlou
- Istanbul esenyurt University, Physical Education and Sports High School, Coaching Education Department, Istanbul, Turkey
| | - Alireza Norouzi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | | |
Collapse
|
11
|
Dai X, Feng S, Li T. Cold atmospheric plasma control metabolic syndromes via targeting fat mass and obesity-associated protein. Pharmacol Res 2025; 215:107720. [PMID: 40174815 DOI: 10.1016/j.phrs.2025.107720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Both obesity and metabolic disorders are global medical problems. Driven by prolonged inflammation, obesity increases the risk of developing metabolic syndromes such as fatty liver, diabetes, cardiovascular diseases and cancers. The fat mass and obesity-associated protein (FTO) is an m6A demethylase, elevated activity of which is known to promote the pathogenesis of many metabolic disorders, leading to the establishment of various FTO inhibitors. By combing through intrinsic connections among obesity and the four primary metabolic problems, we attribute their shared pathological cause to prolonged inflammation. By reviewing the roles of FTO in promoting these disorders and the current status of existing FTO inhibitors in treating these syndromes, we underpinned the paramount potential of resolving these clinical issues by targeting FTO and the urgent need of establishing novel FTO inhibitors with maximized efficacy and minimized side effect. Cold atmospheric plasma (CAP) is the fourth state of matter with demonstrated efficacy in treating various diseases associated with chronic inflammation. By introducing the medical characteristics of CAP, we proposed it as a possible solution to unresolved issues of current FTO inhibitors given its anti-inflammation feature and demonstrated clinical safety. We also emphasized the need of intensive investigations in exploring the feasibility of using CAP in treating obesity and associated metabolic syndromes that might function through targeting FTO.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| | - Shuo Feng
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Tian Li
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, 8 Changjiang Avenue, Tianjin 300100, China.
| |
Collapse
|
12
|
Yen FS, Wei JCC, Wang C, Hou MC, Yu TS, Huang Y, Hwu CM, Hsu CC. Sodium-Glucose Cotransporter2 inhibitors and associated Liver-Related outcomes in diabetes patients. Diabetes Res Clin Pract 2025; 223:112174. [PMID: 40228671 DOI: 10.1016/j.diabres.2025.112174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is associated with poorer liver-related outcomes in type 2 diabetes (T2D) patients. We compared risk of liver-related outcomes and all-cause mortality between sodium-glucose cotransporter-2 inhibitor (SGLT-2i) users and nonusers in T2D patients. METHODS We identified 282,161 T2D patients from the Taiwan National Health Insurance Research Database (2009-2020), excluding those with viral hepatitis or alcohol-related disorders. Propensity score matching created patient pairs on SGLT-2i and other antidiabetic drugs, and Cox proportional hazard models assessed outcomes. RESULTS Over a mean follow-up of 2.7 years, SGLT-2i use was associated with significantly lower risk of liver cirrhosis (adjusted hazard ratio [aHR] 0.78, 95 % CI: 0.70-0.87), decompensated cirrhosis (aHR 0.79, 95 % CI: 0.70-0.89), liver failure (aHR 0.78, 95 % CI: 0.68-0.89), and all-cause mortality (aHR 0.52, 95 % CI: 0.51-0.54). Compared to dipeptidyl peptidase-4 inhibitors (DPP-4i), glucagon-like peptide-1 receptor agonists (GLP-1 RA), pioglitazone, and sulfonylureas, SGLT-2i use was associated with lower risk of cirrhosis, liver failure, and all-cause mortality. SGLT-2i use was associated with lower liver-related mortality than DPP-4i or GLP-1 RA use. CONCLUSIONS This cohort study suggests that SGLT-2i may reduce the risk of liver cirrhosis, decompensated cirrhosis, liver failure, and liver-related and all-cause mortality in T2D patients.
Collapse
Affiliation(s)
| | - James Cheng-Chung Wei
- Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chen Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Teng-Shun Yu
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung City, Taiwan
| | - Yuhan Huang
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan; College of Medicine, China Medical University, Taichung City, Taiwan
| | - Chii-Min Hwu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan; Section of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; National Center for Geriatrics and Welfare Research, National Health Research Institutes, Yunlin, Taiwan.
| |
Collapse
|
13
|
Huang Q, Qadri SF, Bian H, Yi X, Lin C, Yang X, Zhu X, Lin H, Yan H, Chang X, Sun X, Ma S, Wu Q, Zeng H, Hu X, Zheng Y, Yki-Järvinen H, Gao X, Tang H, Xia M. A metabolome-derived score predicts metabolic dysfunction-associated steatohepatitis and mortality from liver disease. J Hepatol 2025; 82:781-793. [PMID: 39423864 DOI: 10.1016/j.jhep.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/11/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) is associated with a >10-fold increase in liver-related mortality. However, biomarkers predicting both MASH and mortality in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) are missing. We developed a metabolome-derived prediction score for MASH and examined whether it predicts mortality in Chinese and European cohorts. METHODS The MASH prediction score was developed using a multi-step machine learning strategy, based on 44 clinical parameters and 250 serum metabolites measured by proton nuclear magnetic resonance in 311 Chinese adults undergoing a liver biopsy. External validation was conducted in a Finnish liver biopsy cohort (n = 305). We investigated associations of the score with all-cause and cause-specific mortality in the population-based Shanghai Changfeng study (n = 5,893) and the UK biobank (n = 111,673). RESULTS A total of 24 clinical parameters and 194 serum metabolites were significantly associated with MASH in the Chinese liver biopsy cohort. The final MASH score included BMI, aspartate aminotransferase, tyrosine, and the phospholipid-to-total lipid ratio in VLDL. The score identified patients with MASH with AUROCs of 0.87 (95% CI 0.83-0.91) and 0.81 (95% CI 0.75-0.88) in the Chinese and Finnish cohorts, with high negative predictive values. Participants with a high or intermediate risk of MASH based on the score had a markedly higher risk of MASLD-related mortality than those with a low risk in Chinese (hazard ratio 23.19; 95% CI 4.80-111.97) and European (hazard ratio 20.15; 95% CI 10.95-37.11) individuals after 7.2 and 12.6 years of follow-up, respectively. The MASH prediction score was superior to the Fibrosis-4 index and the NAFLD fibrosis score in predicting MASLD-related mortality. CONCLUSION The metabolome-derived MASH prediction score accurately predicts risk of MASH and MASLD-related mortality in both Chinese and European individuals. IMPACT AND IMPLICATIONS Metabolic dysfunction-associated steatohepatitis (MASH) is associated with more than a 10-fold increase in liver-related death. However, biomarkers predicting not only MASH, but also death due to liver disease, are missing. We established a MASH prediction score based on 44 clinical parameters and 250 serum metabolites using a machine learning strategy. This metabolome-derived MASH prediction score could accurately identify patients with MASH among both Chinese and Finnish individuals, and it was superior to the Fibrosis-4 index and the NAFLD fibrosis score in predicting MASLD-related death in the general population. Thus, the new MASH prediction score is a useful tool for identifying individuals with a markedly increased risk of serious liver-related outcomes among at-risk and general populations.
Collapse
Affiliation(s)
- Qingxia Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Sami F Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiaoxuan Yi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Chenhao Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinyu Yang
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiaopeng Zhu
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xinxia Chang
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiaoyang Sun
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Shuai Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Qi Wu
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Hailuan Zeng
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiqi Hu
- Department of Pathology, Medical College, Fudan University, Shanghai, China
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China.
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Zhao E, Xie H, Gao Y, Wen X, Dong B, Zhang C. Association Between High Sensitivity Cardiac Troponin and All-Cause and Cardiovascular Mortality in Adults at Risk of Non-Alcoholic Fatty Liver Disease: A Cohort Study. Glob Heart 2025; 20:40. [PMID: 40322053 PMCID: PMC12047645 DOI: 10.5334/gh.1427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Objective Cardiovascular disease (CVD) is the leading cause of death among patients with non-alcoholic fatty liver disease (NAFLD). This study investigates the association between high-sensitivity cardiac troponin (hs-cTn) levels and mortality in adults at risk of NAFLD in a representative U.S. population sample. Methods Among participants aged 18 years and older in the 1999-2004 National Health and Nutrition Examination Survey, we measured high-sensitivity troponin T using a single assay (Roche) and high-sensitivity troponin I using three assays (Abbott, Siemens, and Ortho). Myocardial injury was identified by elevated levels of hs-cTn. Mortality outcomes were determined through linkage with the National Death Index database, with follow-up until December 31, 2019. A multivariable Cox proportional hazards model was used to evaluate the associations between myocardial injury and mortality in the NAFLD population. Sensitivity analyses were conducted to assess the robustness of the main findings. Results A total of 2581 at risk of NAFLD were included in this observational study, with myocardial injury identified in 7.01%. Over a median follow-up of 16.7 years, 937 all-cause deaths occurred, including 319 cardiovascular disease-related deaths. NAFLD individuals with myocardial injury had worse survival rates at 5, 10, and 15 years compared to those without myocardial injury. After adjusting for baseline characteristics, myocardial injury was associated with an increased risk of all-cause mortality (adjusted Hazard Ratio [aHR] 1.785, 95% CI 1.494-2.134, P < 0.001) and cardiovascular mortality (aHR 2.155, 95% CI 1.606-2.893, P < 0.001). Conclusion This large, nationally representative study demonstrates that myocardial injury, defined by elevated hs-cTn levels, is independently associated with increased all-cause and cardiovascular mortality risks in the adult population at risk of NAFLD in the United States. This association persisted after adjusting for various factors and in patients without pre-existing cardiovascular disease. The Siemens hs-cTn I assay demonstrated the strongest association with all-cause mortality. These findings highlight the potential of hs-cTn as a valuable prognostic marker in NAFLD patients, even in those without clinically apparent cardiovascular disease. Routine hs-cTn assessment may aid in risk stratification and guide targeted interventions to reduce mortality risk in this population.
Collapse
Affiliation(s)
- Enfa Zhao
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Hang Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Yuan Gao
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Xiaolin Wen
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Bingtian Dong
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Chaoxue Zhang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
15
|
Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Kunduzova O, Oksenych V, Kamyshnyi O. Unlocking the gut-liver axis: microbial contributions to the pathogenesis of metabolic-associated fatty liver disease. Front Microbiol 2025; 16:1577724. [PMID: 40351307 PMCID: PMC12061941 DOI: 10.3389/fmicb.2025.1577724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex metabolic disorder characterized by hepatic lipid accumulation and subsequent inflammation. This condition is closely linked to metabolic syndrome and obesity, with its prevalence rising due to sedentary lifestyles and high-calorie diets. The pathogenesis of MAFLD involves multiple factors, including insulin resistance, lipotoxicity, oxidative stress, and inflammatory responses. The gut microbiota plays a crucial role in MAFLD development, with dysbiosis contributing to liver inflammation through various mechanisms, such as enhanced intestinal permeability and the translocation of bacterial products like lipopolysaccharide (LPS). Microbial metabolites, including short-chain fatty acids (SCFAs) and bile acids, influence hepatic function and immune responses, with potential implications for disease progression. Specific gut microbiome signatures have been identified in MAFLD patients, offering potential diagnostic and therapeutic targets. Moreover, gut-derived toxins, such as endotoxins, lipopolysaccharides, trimethylamine-N-oxide and bacterial metabolites, significantly influence liver damage and inflammation, highlighting the complex interplay between the gut microbiome and hepatic health. This review comprehensively examines the complex interplay between the gut microbiota and MAFLD, focusing on underlying pathogenic mechanisms, potential biomarkers, and emerging microbiome-targeted therapeutic strategies for disease management.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University, Toulouse, France
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
16
|
Iakovleva V, de Jong YP. Gene-based therapies for steatotic liver disease. Mol Ther 2025:S1525-0016(25)00298-9. [PMID: 40254880 DOI: 10.1016/j.ymthe.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Advances in nucleic acid delivery have positioned the liver as a key target for gene therapy, with adeno-associated virus vectors showing long-term effectiveness in treating hemophilia. Steatotic liver disease (SLD), the most common liver condition globally, primarily results from metabolic dysfunction-associated and alcohol-associated liver diseases. In some individuals, SLD progresses from simple steatosis to steatohepatitis, cirrhosis, and eventually hepatocellular carcinoma, driven by a complex interplay of genetic, metabolic, and environmental factors. Genetic variations in various lipid metabolism-related genes, such as patatin-like phospholipase domain-containing protein 3 (PNPLA3), 17β-hydroxysteroid dehydrogenase type 13 (HSD17B13), and mitochondrial amidoxime-reducing component 1 (MTARC1), impact the progression of SLD and offer promising therapeutic targets. This review largely focuses on genes identified through clinical association studies, as they are more likely to be effective and safe for therapeutic intervention. While preclinical research continues to deepen our understanding of genetic factors, early-stage clinical trials involving gene-based SLD therapies, including transient antisense and small-molecule approaches, are helping prioritize therapeutic targets. Meanwhile, hepatocyte gene editing technologies are advancing rapidly, offering alternatives to transient methods. As such, gene-based therapies show significant potential for preventing the progression of SLD and enhancing long-term liver health.
Collapse
Affiliation(s)
- Viktoriia Iakovleva
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
17
|
Giannakodimos A, Oikonomou E, Pantelidis P, Theofilis P, Katsiki N, Goliopoulou A, Zakynthinos GE, Korakas E, Kalogera V, Banach M, Lampadiari V, Kassi E, Ikonomidis I, Siasos G. Arterial stiffness as a complication of metabolic dysfunction-associated steatotic liver disease: a systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol 2025; 19:413-426. [PMID: 39988816 DOI: 10.1080/17474124.2025.2471871] [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: 12/22/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION The purpose of this systematic review and meta-analysis is to investigate the association of metabolic dysfunction-associated steatotic liver disease (MASLD) with arterial stiffness and enlighten on potential cardiometabolic co-factors. METHODS A literature search in PubMed/Medline, Embase, Scopus, and Web of Science databases was conducted. All the observational studies comparing arterial stiffness indices between adults with Non-alcoholic Fatty Liver Disease (NAFLD), Metabolic Dysfunction Associated-Fatty Liver Disease (MAFLD), or MASLD and apparently healthy individuals with normal liver function were included. Pulse wave velocity (PWV) and augmentation index (AIx) were mainly used as arterial stiffness indices. RESULTS Fourty one unique studies were included in the systematic review, with 27 deemed eligible for meta-analysis. Patients with MASLD had increased carotid-femoral PWV (14 studies, Mean difference (MD): 0.96 m/s, 95% confidence interval (CI) 0.65-1.27, p < 0.001) compared with healthy individuals. This finding was independent from body mass index, triglycerides, high-density lipoprotein, systolic blood pressure, and fasting plasma glucose. Moreover, patients with MASLD had higher brachial-ankle PWV (13 studies, MD: 78.14 cm/s, 95% CI 60.37-95.90, p < 0.001) and AIx (7 studies, MD: 3.85%, 95% CI 0.87-6.82, p = 0.0195) compared with controls. CONCLUSIONS MASLD is correlated with increased arterial stiffness. This relation is unaffected by common cardiometabolic risk factors. REGISTRATION PROSPERO (ID: CRD42023468258).
Collapse
Affiliation(s)
- Alexios Giannakodimos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Athina Goliopoulou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios E Zakynthinos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanouil Korakas
- 2nd Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Kalogera
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vaia Lampadiari
- 2nd Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ignatios Ikonomidis
- 2nd Cardiology Department, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
18
|
Dixon ED, Claudel T, Nardo AD, Riva A, Fuchs CD, Mlitz V, Busslinger G, Scharnagl H, Stojakovic T, Senéca J, Hinteregger H, Grabner GF, Kratky D, Verkade H, Zimmermann R, Haemmerle G, Trauner M. Inhibition of ATGL alleviates MASH via impaired PPARα signalling that favours hydrophilic bile acid composition in mice. J Hepatol 2025; 82:658-675. [PMID: 39357546 DOI: 10.1016/j.jhep.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND & AIMS Adipose triglyceride lipase (ATGL) is an attractive therapeutic target in insulin resistance and metabolic dysfunction-associated steatotic liver disease (MASLD). This study investigated the effects of pharmacological ATGL inhibition on the development of metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis in mice. METHODS Streptozotocin-injected male mice were fed a high-fat diet to induce MASH. Mice receiving the ATGL inhibitor atglistatin (ATGLi) were compared to controls using liver histology, lipidomics, metabolomics, 16s rRNA, and RNA sequencing. Human ileal organoids, HepG2 cells, and Caco2 cells treated with the human ATGL inhibitor NG-497, HepG2 ATGL knockdown cells, gel-shift, and luciferase assays were analysed for mechanistic insights. We validated the benefits of ATGLi on steatohepatitis and fibrosis in a low-methionine choline-deficient mouse model. RESULTS ATGLi improved serum liver enzymes, hepatic lipid content, and histological liver injury. Mechanistically, ATGLi attenuated PPARα signalling, favouring hydrophilic bile acid (BA) synthesis with increased Cyp7a1, Cyp27a1, Cyp2c70, and reduced Cyp8b1 expression. Additionally, reduced intestinal Cd36 and Abca1, along with increased Abcg5 expression, were consistent with reduced levels of hepatic triacylglycerol species containing polyunsaturated fatty acids, like linoleic acid, as well as reduced cholesterol levels in the liver and plasma. Similar changes in gene expression associated with PPARα signalling and intestinal lipid transport were observed in ileal organoids treated with NG-497. Furthermore, HepG2 ATGL knockdown cells revealed reduced expression of PPARα target genes and upregulation of genes involved in hydrophilic BA synthesis, consistent with reduced PPARα binding and luciferase activity in the presence of the ATGL inhibitors. CONCLUSIONS Inhibition of ATGL attenuates PPARα signalling, translating into hydrophilic BA composition, interfering with dietary lipid absorption, and improving metabolic disturbances. Validation with NG-497 opens a new therapeutic perspective for MASLD. IMPACT AND IMPLICATIONS Despite the recent approval of drugs novel mechanistic insights and pathophysiology-oriented therapeutic options for MASLD (metabolic dysfunction-associated steatotic liver disease) are still urgently needed. Herein, we show that pharmacological inhibition of ATGL, the key enzyme in lipid hydrolysis, using atglistatin (ATGLi), improves MASH (metabolic dysfunction-associated steatohepatitis), fibrosis, and key features of metabolic dysfunction in mouse models of MASH and liver fibrosis. Mechanistically, we demonstrated that attenuation of PPARα signalling in the liver and gut favours hydrophilic bile acid composition, ultimately interfering with dietary lipid absorption. One of the drawbacks of ATGLi is its lack of efficacy against human ATGL, thus limiting its clinical applicability. Against this backdrop, we could show that ATGL inhibition using the human inhibitor NG-497 in human primary ileum-derived organoids, Caco2 cells, and HepG2 cells translated into therapeutic mechanisms similar to ATGLi. Collectively, these findings reveal a possible new avenue for MASLD treatment.
Collapse
Affiliation(s)
- Emmanuel Dauda Dixon
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Alexander Daniel Nardo
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Alessandra Riva
- Chair of Nutrition and Immunology, School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Claudia Daniela Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Veronika Mlitz
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Georg Busslinger
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Tatjana Stojakovic
- Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Austria
| | - Joana Senéca
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria; Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Helga Hinteregger
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Gernot F Grabner
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Dagmar Kratky
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Henkjan Verkade
- Department of Paediatrics, University Medical Centre Groningen, Groningen, Netherlands
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Basile L, Cannarella R, Magni P, Condorelli RA, Calogero AE, La Vignera S. Role of gliflozins on hepatocellular carcinoma progression: a systematic synthesis of preclinical and clinical evidence. Expert Opin Drug Saf 2025; 24:413-426. [PMID: 39714931 DOI: 10.1080/14740338.2024.2447057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/02/2024] [Accepted: 12/22/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION The risk of HCC is twice as high in diabetic patients compared to non-diabetic ones, suggesting that diabetes advances carcinogenesis in the liver through a variety of mechanisms. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have been shown to improve liver outcomes, emerging as promising agents to treat hepatocellular carcinoma (HCC) in patients with type 2 diabetes mellitus (T2DM). METHODS We searched PubMed and Scopus databases for articles presenting an association between SGLT2is and HCC to explore the putative mechanisms of action underlying the anti-proliferative activity of SGLT2is. RESULTS A total of 24 articles were selected for inclusion, of which 14 were preclinical and 10 were clinical. Preclinical studies were mainly focused on canagliflozin, used alone or in combination with other drugs. CONCLUSIONS Overall, canagliflozin had a negative effect on HCC cell proliferation by interfering with glucose-dependent and independent metabolic pathways, negatively impacting angiogenesis, and inducing apoptosis in in-vitro cell models. In-vivo, a protective effect on hepatic steatosis and fibrosis and HCC development has been reported. Human studies showed a lower risk of developing HCC in patients on SGLT2is. However, this is supported by retrospective cohort studies. Clinical trials are needed to confirm the causal relationship between SGLT2i administration and HCC development.
Collapse
Affiliation(s)
- Livia Basile
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
20
|
Liu Z, Peng H, Liu P, Duan F, Yang Y, Li P, Li Z, Wu J, Chang J, Shang D, Tian Q, Zhang J, Xie Y, Liu Z, An Y. Deciphering significances of autophagy in the development and metabolism of adipose tissue. Exp Cell Res 2025; 446:114478. [PMID: 39978716 DOI: 10.1016/j.yexcr.2025.114478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
The mechanisms of adipose tissue activation and inactivation have been a hot topic of research in the last decade, from which countermeasures have been attempted to be found against obesity as well as other lipid metabolism-related diseases, such as type 2 diabetes mellitus and non-alcoholic fatty liver disease. Autophagy has been shown to be closely related to the regulation of adipocyte activity, which is involved in the whole process including white adipocyte differentiation/maturation and brown or beige adipocyte generation/activation. Dysregulation of autophagy in adipose tissue has been demonstrated to be associated with obesity. On this basis, we summarize the pathways and mechanisms of autophagy involved in the regulation of lipid metabolism and present a review of its pathophysiological roles in lipid metabolism-related diseases, in the hope of providing ideas for the treatment of these diseases.
Collapse
Affiliation(s)
- Zitao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Haoyuan Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengfei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Feiyi Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yutian Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiaoyan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiayi Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Dandan Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Qiwen Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Jiawei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yucheng Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Zhenzhen Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China; Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 450064, China.
| |
Collapse
|
21
|
Misra A, Kumar A, Kuchay MS, Ghosh A, Gulati S, Choudhary NS, Dutta D, Sharma P, Vikram NK. Consensus guidelines for the diagnosis and management of metabolic dysfunction-associated steatotic liver disease in adult Asian Indians with type 2 diabetes. Diabetes Metab Syndr 2025; 19:103209. [PMID: 40222341 DOI: 10.1016/j.dsx.2025.103209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 04/15/2025]
Affiliation(s)
- Anoop Misra
- Fortis CDOC Center of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India; National Diabetes Obesity and Cholesterol Foundation (N-DOC), New Delhi, India; Diabetes Foundation India, New Delhi, India.
| | - Ashish Kumar
- Gastroenterology & Hepatology,Sir Ganga Ram Hospital, Rajinder Nagar New Delhi, India
| | - Mohammad Shafi Kuchay
- Division of Endocrinology and Diabetes, Medanta, The Medicity, Gurugram, 122001, Haryana, India
| | - Amerta Ghosh
- Fortis CDOC Center of Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India; National Diabetes Obesity and Cholesterol Foundation (N-DOC), New Delhi, India
| | - Seema Gulati
- National Diabetes Obesity and Cholesterol Foundation (N-DOC), New Delhi, India; Diabetes Foundation India, New Delhi, India
| | | | - Deep Dutta
- Department of Endocrinology, Center for Endocrinology, Diabetes, Arthritis & Rheumatism (CEDAR) Super speciality Clinics, New Delhi, India
| | - Praveen Sharma
- Gastroenterology & Hepatology,Sir Ganga Ram Hospital, Rajinder Nagar New Delhi, India
| | - Naval K Vikram
- Department of Internal Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| |
Collapse
|
22
|
Hong H, Fu Q, Gu P, Zhao J, Dai J, Xu K, Yang T, Dai H, Shen S. Investigating the common genetic architecture and causality of metabolic disorders with neurodegenerative diseases. Diabetes Obes Metab 2025; 27:1337-1349. [PMID: 39703124 DOI: 10.1111/dom.16130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The co-occurrence of metabolic dysfunction and neurodegenerative diseases suggests a genetic link, yet the shared genetic architecture and causality remain unclear. We aimed to comprehensively characterise these genetic relationships. METHODS We investigated genetic correlations among four neurodegenerative diseases and seven metabolic dysfunctions, followed by bidirectional Mendelian randomisation (MR) to assess potential causal relationships. Pleiotropy analysis (PLACO) was used to detect the pleiotropic effects of genetic variants. Significant pleiotropic loci were refined and annotated using functional mapping and annotation (FUMA) and Bayesian colocalisation analysis. We further explored mapped genes with tissue-specific expression and gene set enrichment analyses. RESULTS We identified significant genetic correlations in nine out of 28 trait pairs. MR suggested causal relationships between specific trait pairs. Pleiotropy analysis revealed 25 931 significant single-nucleotide polymorphisms, with 246 pleiotropic loci identified via FUMA and 55 causal loci through Bayesian colocalisation. These loci are involved in neurotransmitter transport and immune response mechanisms, notably the missense variant rs41286192 in SLC18B1. The tissue-specific analysis highlighted the pancreas, left ventricle, amygdala, and liver as critical organs in disease progression. Drug target analysis linked 74 unique genes to existing therapeutic agents, while gene set enrichment identified 189 pathways related to lipid metabolism, cell differentiation and immune responses. CONCLUSION Our findings reveal a shared genetic basis, pleiotropic loci, and potential causal relationships between metabolic dysfunction and neurodegenerative diseases. These insights highlight the biological connections underlying their phenotypic association and offer implications for future research to reduce the risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hao Hong
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qi Fu
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pan Gu
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingyi Zhao
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jinglan Dai
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Kuanfeng Xu
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Yang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Dai
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Sipeng Shen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Brito CF, Fonseca RC, Rodrigues-Ribeiro L, Guimarães JSF, Vaz BF, Tofani GSS, Batista ACS, Diniz AB, Fernandes P, Nunes NAM, Pessoa RM, Oliveira ACC, Lula IS, Cardoso VN, Fernandes SOA, Poletini MO, Alvarez-Leite JI, Menezes GB, Ferreira AVM, Magalhães MTQ, Gorshkov V, Kjeldsen F, Verano-Braga T, Araujo AM, Oliveira AG. Vagus Nerve Mediated Liver-Brain-Axis Is a Major Regulator of the Metabolic Landscape in the Liver. Int J Mol Sci 2025; 26:2166. [PMID: 40076796 PMCID: PMC11901116 DOI: 10.3390/ijms26052166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The liver serves as a major energetic reservoir for other tissues and its metabolic function is controlled by humoral and neural factors. The vagus nerve innervating the gastrointestinal tract plays an important role in regulating peripheral metabolism and energy expenditure. Although the liver receives vagus nerve fibers, the impact of this circuitry in the regulation of hepatic metabolism is still poorly understood. Herein, we used a combination of quantitative proteomics and in vivo imaging techniques to investigate the impact of the vagus nerve on liver metabolism in male mice. Liver-brain axis was impaired by vagotomy (VNX) or knocking down of the vesicular acetylcholine transporter (VAChT-KD). Mice were challenged with high carbohydrate or high-fat feeding. The vagus nerve shapes the metabolic framework of the liver, as vagotomy led to a significant alteration of the hepatic proteome landscape. Differential protein expression and pathway enrichment analyses showed that glycolytic and fatty acid biosynthesis were increased following VNX, whereas β-oxidation was decreased. These results were corroborated in VAChT-KD mice. This metabolic shift facilitated lipid accumulation in hepatocytes in mice fed with a standard commercial diet. Furthermore, VNX worsened liver steatosis following high-carbohydrate or high-fat dietary challenges. This study describes the liver-brain axis mediated by the vagus nerve as an important regulator of the hepatic metabolic landscape.
Collapse
Affiliation(s)
- Camila F. Brito
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Roberta C. Fonseca
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Lucas Rodrigues-Ribeiro
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - João S. F. Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Bruna F. Vaz
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Gabriel S. S. Tofani
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Ana C. S. Batista
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Ariane B. Diniz
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Paola Fernandes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Núbia A. M. Nunes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Rafaela M. Pessoa
- Department of Clinical and Toxicological Analysis, College of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Amanda C. C. Oliveira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Ivana S. Lula
- Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Valbert N. Cardoso
- Department of Clinical and Toxicological Analysis, College of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Simone O. A. Fernandes
- Department of Clinical and Toxicological Analysis, College of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Maristela O. Poletini
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Jacqueline I. Alvarez-Leite
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Gustavo B. Menezes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Adaliene V. M. Ferreira
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Mariana T. Q. Magalhães
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vladimir Gorshkov
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Frank Kjeldsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Thiago Verano-Braga
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| | - Alan M. Araujo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA
| | - André G. Oliveira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (C.F.B.); (R.C.F.)
| |
Collapse
|
24
|
Wang H, Zheng C, Wang P. Exploring the nexus between hypothyroidism and metabolic dysfunction-associated steatotic liver disease: a UK biobank cohort study. Sci Rep 2025; 15:6692. [PMID: 40000892 PMCID: PMC11862249 DOI: 10.1038/s41598-025-91221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease characterised by lipid deposition in liver cells. The global prevalence of NAFLD has significantly increased from 8.2% in 1990 to 30.2% in 2023, establishing it as a growing public health concern. In recent years, the name NAFLD has been replaced by metabolic dysfunction-associated fatty liver disease (MASLD). Numerous observational studies have investigated the potential association between hypothyroidism and MASLD; however, the findings remain inconsistent. In this context, a systematic analysis was conducted to examine the relationship between hypothyroidism and MASLD using data from a large cohort within the UK Biobank. Utilising prospective data from the UK Biobank, a Cox proportional hazards model supplemented with multiple sensitivity analyses was applied to investigate the association between the incidence of hypothyroidism and the onset of MASLD. In addition, stratified analyses and prognostic assessments were performed to assess potential effect modifiers. To explore the underlying mechanisms, mediation analyses were conducted, along with restricted cubic spline regression, to examine potential non-linear relationships and mediation effects within this association. The study found that after fully adjusting for multiple covariates, the risk of MASLD in hypothyroidism patients was 1.711 times that of non-hypothyroidism patients (95% CI 1.560-1.877, P < 0.001). Both subtypes of hypothyroidism, namely non-surgical related hypothyroidism (NSRH) and surgical related hypothyroidism (SRH), were associated with a markedly elevated risk of MASLD onset. For NSRH, the risk is increased by 1.710 times (95% CI 1.557-1.878, P < 0.001), and for SRH, the risk is increased by 1.763 times (95% CI 1.344-2.313, P < 0.001). Stratified analysis revealed an interaction effect between gender and BMI in relation to the risk of MASLD among individuals with NSRH. Mediation analysis revealed the critical role of specific biomarkers in elucidating the relationship between hypothyroidism and MASLD. Notably, red cell distribution width, C-reactive protein, HbA1c, and total protein were identified as significant mediators in this association. Patients with hypothyroidism exhibit a significantly increased risk of developing MASLD, with inflammatory and metabolic markers playing a mediating role in this association. These findings suggest that individuals with hypothyroidism, particularly those with elevated levels of inflammatory markers, may be at heightened risk for MASLD. As such, enhanced clinical monitoring of liver function in these patients is recommended to facilitate early detection and intervention.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun, China
| | - Changlin Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Lequn Branch, The First Hospital of Jilin University, Changchun, China
| | - Peisong Wang
- Thyroid Surgery Department, General Surgery Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
25
|
He Q, Liu X, Ding G, Wang Y, Luo X, Cao W, Xing W. The relationship between serum uric acid level and non-alcoholic fatty liver disease in northern China: a retrospective cohort study. BMC Public Health 2025; 25:718. [PMID: 39984884 PMCID: PMC11843771 DOI: 10.1186/s12889-025-21943-w] [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: 09/28/2023] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease among adults. High uric acid (UA) increases the incidence of NAFLD in the general population. However, further exploration is warranted to determine the relationship between UA levels and NAFLD in various populations. We conducted a historical cohort study to investigate the causality between UA and NAFLD across different weight categories. METHODS A historical cohort was established from the Jidong community cohort. All participants were enrolled and followed up from July 1st, 2013 to August 1st, 2018. The study participants were retrospectively assigned to four groups according to their UA levels (Q1, 69-210 μmol/L; Q2, 211-255 μmol/L; Q3, 256-310 μmol/L; Q4, 311-593 μmol/L). The NAFLD incidence was investigated in each group. We used the UA level determined by an automatic analyzer. NAFLD was diagnosed with abdominal ultrasonography examination. Demographic information, lifestyle history, clinical anthropometric data, and blood samples of participants were collected. Univariate analysis and multivariable Cox regression were applied to analyze the relationship between UA and NAFLD by stratification of participants' body mass index (BMI) categories (underweight, normal weight, overweight, and obese). RESULTS Two thousand nine hundred eighty four participants were enrolled. 740 (24.8%) were assigned to UA Q1 group, 755 (25.3%) to UA Q2, 743 (24.9%) to UA Q3, and 746 (25.0%) to UA Q4. The global incidence of NAFLD was 26.0% (777/2984). The risk of NAFLD significantly increased with elevated UA levels in underweight and normal-weight participants (HR = 3.498, 95% CI: 2.413-5.072, P < 0.05). In multivariable analysis, UA showed a positive association with NAFLD, independent of other risk factors in underweight and normal-weight participants (UA Q2: 1.152 (0.761-1.743), UA Q3: 2.168 (1.489-3.157), UA Q4: 3.075 (2.103-4.196), P < 0.05). In the absence of other risk factors, high UA levels independently explained 17% of NAFLD risk in underweight and normal-weight participants. CONCLUSIONS High UA levels serve as an independent risk factor for NAFLD in underweight and normal-weight individuals, highlighting the necessity of early NAFLD screening through monitoring liver function and UA levels, and personalized treatment plans for NAFLD patients with higher UA levels, which may include uric acid-lowering therapy and lifestyle modifications. However, the relationship between UA levels and NAFLD in overweight and obese individuals remains inconclusive.
Collapse
Affiliation(s)
- Qian He
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xinyue Liu
- Tai'an City Center for Disease Control and Prevention, Taian, China
| | - Guoyong Ding
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yiying Wang
- Department of Medical, Rizhao Mental Health Center, Rizhao, China
| | - Xiaoting Luo
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wenyuan Cao
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weijia Xing
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, 271000, China.
| |
Collapse
|
26
|
Zheng Y, Sun Y, Ren W, Duan R, Li S, Chen M, Qin H, Ying M, Ren J. Factors Associated with Nonalcoholic Fatty Liver Disease in a Non-Overweight/Obese and Overweight/Obese Chinese Population at Risk for Metabolic Syndrome: A Cross-Sectional Multicenter Study. Metab Syndr Relat Disord 2025; 23:41-52. [PMID: 39311687 DOI: 10.1089/met.2024.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
Background: To investigate the association of demographic, clinical, and metabolic factors with nonalcoholic fatty liver disease (NAFLD) in a non-overweight/obese and overweight/obese Chinese population at risk for metabolic syndrome. Patients and Method: A cross-sectional multicenter study was conducted using convenience sampling from eight selected counties/cities in Zhejiang, China, between May 2021 and September 2022. Demographics, epidemiological, anthropometric, and clinical characteristics were obtained from a questionnaire. Least absolute shrinkage and selection operator (LASSO)-logistic regression analysis was used to identify the variables associated with NAFLD. Receiver operating characteristic (ROC) curve analysis and decision curve analysis (DCA) were performed to evaluate the diagnostic value and clinical utility of the variables and models. Results: A total of 1739 patients were enrolled in the final analysis, 345 (19.8%) were non-overweight/obese and 1394 (80.2%) were overweight/obese participants. There were 114 (33.0%) and 1094 (78.5%) patients who met the criteria for NAFLD in the non-overweight/obese participants and the overweight/obese participants respectively. Older age, current smoking, higher triglyceride (TG) levels, higher AST levels, higher albumin levels, lower insulin levels, and higher controlled attenuation parameter (CAP) scores were associated with NAFLD in both non-overweight/obese and overweight/obese participants. The combination of TG+CAP scores had strong predictive values for NAFLD, especially in non-overweight/obese (Area Under Curve = 0.812, 95% confidence interval: 0.764-0.863). DCA showed a superior net benefit of the TG+CAP score over other variables or models, suggesting a better clinical utility in identifying NAFLD. Conclusions: More stringent lipid management strategies remain essential, and the convenience and efficacy of transient elastography for liver steatosis should be recognized, especially in the non-overweight/obese population.
Collapse
Affiliation(s)
- Yang Zheng
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Allergy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yujing Sun
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen Ren
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ruoshu Duan
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Li
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mingmin Chen
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongli Qin
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Meike Ying
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Ren
- Department of General Practice, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Zhu L, Li Y, Yu X, Chen Y, Zhang J, Pang C, Xie J, Gao L, Du L, Cao W, Fan D, Cui C, Yu H, Deng B. Fighting Amyotrophic Lateral Sclerosis by Protecting the Liver? A Prospective Cohort Study. Ann Neurol 2025; 97:270-280. [PMID: 39425590 DOI: 10.1002/ana.27115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Previous studies have observed liver abnormalities in amyotrophic lateral sclerosis (ALS) patients. This study aimed to investigate whether early signs of liver disease, measured by magnetic resonance imaging-derived iron-corrected T1-mapping (cT1), are risk factors for developing ALS. METHODS cT1 and proton density fat fraction were measured and automatically analyzed using LiverMultiScan® software. The Fibrosis-4 index was calculated using an established formula based on age and blood markers. Cox proportional hazard models were used to examine the relationship between liver disease, liver biomarkers, and incident ALS. RESULTS In a cohort of 533,707 individuals from UK Biobank, 24 ALS cases were identified among 28,328 participants with liver disease during the follow-up period. Among a total of 33,959 individuals with complete liver imaging data, 15 incident ALS cases were observed during a median follow-up period of 5.6 years. Individuals with liver disease had a higher risk of developing ALS, with an adjusted hazard ratio of 7.35 (95% CI 4.47-12.09; p < 0.001). An increase in cT1 was also associated with a higher risk of ALS. After adjusting for age, sex, Townsend deprivation index, smoking status, alcohol intake frequency, body mass index, proton density fat fraction, Fibrosis-4, and metabolic syndrome, an increase in cT1 remained significantly associated with a higher risk of ALS, with an adjusted hazard ratio of 3.15 (95% CI 1.79-5.55) per 1-SD increase. Sensitivity analyses confirmed these robust results. INTERPRETATION Liver disease activity, indicated by cT1, increases the risk of developing ALS, independent of metabolic syndrome, liver fat, or fibrosis. ANN NEUROL 2025;97:270-280.
Collapse
Affiliation(s)
- Luyi Zhu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaojia Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyue Yu
- Alberta Institute, Wenzhou Medical University, Wenzhou, China
| | - Yinuo Chen
- First Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Junwei Zhang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunyang Pang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiali Xie
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Neurology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lingfei Gao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lihuai Du
- College of Mathematics and Physics, Wenzhou University, Wenzhou, China
| | - Wen Cao
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Can Cui
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden
| | - Huan Yu
- Department of Pediatrics, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binbin Deng
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
28
|
Athar R, Shahsavan M, Shahabi S, Pazouki A, Husain FA, Kermansaravi M. Impact of Nonalcoholic Fatty Liver Disease on Weight Loss Outcomes After One Anastomosis Gastric Bypass. Surg Laparosc Endosc Percutan Tech 2025; 35:e1347. [PMID: 39588751 DOI: 10.1097/sle.0000000000001347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/10/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Obesity-associated nonalcoholic fatty liver disease (NAFLD) is a significant cause of chronic liver disease. Our study sought to investigate preoperative NAFLD and the effect at 6 months and 2 years after surgery of one anastomosis gastric bypass (OAGB) and its development 6 months after surgery regarding weight loss outcomes. MATERIALS AND METHODS A retrospective cohort study was conducted on patients with severe obesity who underwent primary OAGB at Hazrat-e-Rasool Hospital between March 2020 and June 2021. Preoperative assessments included abdominal ultrasound (US) for NAFLD grading, weight, and biochemical blood tests. Follow-up examinations were performed at 10 days and 1, 3, 6, 9, 12, and 24 months postsurgery, with subsequent US examinations at the 6-month follow-up. RESULTS Two hundred thirty-one patients were included, with an average age of 40.3±10.5 years and a percentage of 78.4 women. Their mean weight and BMI were 131.2±26.8 and 48.8±8.5, respectively. Six-month grades of NAFLD showed that patients with grade 3 NAFLD had significantly lower TWL% compared with the lower grades. NAFLD grades improved in 72.3% of our patients, remained the same at 21.2%, and worsened at 6.5%. The 6-month TWL% was 28.4±4.3 in the no-change group, 28.4±5.3 for the improved group, and 25.2±14.6 in the worse group. CONCLUSION The severity and progression of NAFLD can significantly impact weight loss outcomes post-OAGB, highlighting the importance of monitoring and managing NAFLD in patients undergoing bariatric surgery.
Collapse
Affiliation(s)
- Rahmatullah Athar
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences
| | - Masoumeh Shahsavan
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences
| | - Shahab Shahabi
- Department of Surgery, Minimally Invasive Surgery Research Center, Division of Minimally Invasive and Bariatric Surgery, Hazrat-E Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences
- Center of Excellence of European Branch of International Federation for Surgery of Obesity, Hazrat_e Rasool Hospital, Tehran, Iran
| | - Abdolreza Pazouki
- Department of Surgery, Minimally Invasive Surgery Research Center, Division of Minimally Invasive and Bariatric Surgery, Hazrat-E Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences
- Center of Excellence of European Branch of International Federation for Surgery of Obesity, Hazrat_e Rasool Hospital, Tehran, Iran
| | - Farah A Husain
- Department of Surgery, University of Arizona College of Medicine, Phoenix, AZ
| | - Mohammad Kermansaravi
- Department of Surgery, Minimally Invasive Surgery Research Center, Division of Minimally Invasive and Bariatric Surgery, Hazrat-E Fatemeh Hospital, School of Medicine, Iran University of Medical Sciences
- Center of Excellence of European Branch of International Federation for Surgery of Obesity, Hazrat_e Rasool Hospital, Tehran, Iran
| |
Collapse
|
29
|
Shao W, Xu H, Zeng K, Ye M, Pei R, Wang K. Advances in liver organoids: replicating hepatic complexity for toxicity assessment and disease modeling. Stem Cell Res Ther 2025; 16:27. [PMID: 39865320 PMCID: PMC11771052 DOI: 10.1186/s13287-025-04139-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025] Open
Abstract
The lack of in vivo accurate human liver models hinders the investigation of liver-related diseases, injuries, and drug-related toxicity, posing challenges for both basic research and clinical applications. Traditional cellular and animal models, while widely used, have significant limitations in replicating the liver's complex responses to various stressors. Liver organoids derived from human pluripotent stem cells, adult stem cells primary cells, or tissues can mimic diverse liver cell types, major physiological functions, and architectural features. Recent advancements in the field have shown that some liver organoids have sufficient accuracy to replicate specific aspects of the human liver's complexity. This review highlights recent progress in liver organoid research, with a particular emphasis on their potential for toxicity assessment and disease modeling. The intrinsic advantages of liver organoids include higher sensitivity and suitability for long-term studies, which enhance the predictive value in drug and nanomaterial toxicity testing. The integration of liver organoids with microfluidic devices enables the simulation of the liver microenvironment and facilitates high-throughput drug screening. The liver organoids also serve as ideal platforms for studying liver diseases such as hepatitis, liver fibrosis, viral liver diseases, and monogenic diseases. Additionally, this review discusses the advantages and limitations of liver organoids along with potential avenues for future research.
Collapse
Affiliation(s)
- Weidong Shao
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
- China Pharmaceutical University, 639 Longmian Rd, Nanjing, Jiangsu, 210009, China
| | - Hui Xu
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Kanghua Zeng
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Mingzhou Ye
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Renjun Pei
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China.
| | - Kai Wang
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
30
|
Zhao H, Ji B, Wang X, Shi S, Sheng J, Ma X, Ban B, Gao G. Association between SPISE and NAFLD in patients with type 2 diabetes. Front Med (Lausanne) 2025; 12:1454938. [PMID: 39911865 PMCID: PMC11794499 DOI: 10.3389/fmed.2025.1454938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Aims Non-alcoholic fatty liver disease (NAFLD) is closely related to type 2 diabetes (T2D), with reduced insulin sensitivity being a key factor in their disrupted metabolic processes. The single point insulin sensitivity estimator (SPISE) is a novel index. This study aims to explore the association between SPISE and NAFLD in T2D population. Methods This study included a total of 2,459 patients with T2D. SPISE was calculated based on high density lipoprotein-cholesterol (HDL-c), triglycerides (TG), and body mass index (BMI). Participants were categorized into NAFLD and non-NAFLD groups based on the results of ultrasonographic diagnosis. The relationship between SPISE and NAFLD was analyzed separately for each gender. Results The overall prevalence of NAFLD is 38.5%. In females and males, the SPISE was significantly reduced in the NAFLD group compared to the non-NAFLD group (both P < 0.05). The prevalence of NAFLD showed a significant reduction across quartiles of the SPISE in both genders (both P < 0.05).Additionally, univariate correlation analysis showed a negative correlation between SPISE and NAFLD (both P < 0.05). In multivariate regression analysis, a reduced SPISE was identified as an independent risk factor for NAFLD (odds ratios of 0.572 and 0.737, 95% CI of 0.477-0.687 and 0.587-0.926, respectively).Moreover, the area under the receiver operating characteristic (ROC) curve for SPISE was 0.209 in females and 0.268 in males (95% CI of 0.175-0.244 and 0.216-0.320, respectively). These results are more meaningful than those of other variables. Conclusion SPISE is significantly reduced in NAFLD patients with T2D. Compared to other indicators, SPISE demonstrates superior predictive value in diagnosing NAFLD, and it is independent of gender.
Collapse
Affiliation(s)
- Hongyan Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Endocrinology, Linyi People’s Hospital Affiliated to Shandong Second Medical University, Linyi, Shandong, China
| | - Baolan Ji
- Department of Endocrinology, Linyi People’s Hospital Affiliated to Shandong Second Medical University, Linyi, Shandong, China
| | - Xin Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Shuwei Shi
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Endocrinology, Linyi People’s Hospital Affiliated to Shandong Second Medical University, Linyi, Shandong, China
| | - Jie Sheng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Endocrinology, Linyi People’s Hospital Affiliated to Shandong Second Medical University, Linyi, Shandong, China
| | - Xuan Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Endocrinology, Linyi People’s Hospital Affiliated to Shandong Second Medical University, Linyi, Shandong, China
| | - Bo Ban
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Guanqi Gao
- Department of Endocrinology, Linyi People’s Hospital Affiliated to Shandong Second Medical University, Linyi, Shandong, China
| |
Collapse
|
31
|
Zheng S, Hua T, Yin G, Zhang W, Wang X, Qi L, Jing X, Fan Q, Yu X, Li Y. Metabolic dysfunction-associated fatty liver disease and risk of nephrolithiasis: a sizeable cross-sectional study. Front Endocrinol (Lausanne) 2025; 15:1406065. [PMID: 39906038 PMCID: PMC11790460 DOI: 10.3389/fendo.2024.1406065] [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: 03/24/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
Objective Metabolic dysfunction-associated fatty liver disease (MAFLD) and nephrolithiasis are two common metabolic diseases, but their relationship has not yet been thoroughly studied. Therefore, this study aimed to explore the association between MAFLD and nephrolithiasis and to assess the effect of MAFLD on the risk of nephrolithiasis. Materials and methods This cross-sectional study included 96,767 adults from China. All participants underwent medical examinations, including physical examinations, medical history tests, and laboratory tests. Based on ultrasound examination, participants were divided into MAFLD and non-MAFLD groups, and the severity of liver steatosis was determined based on ultrasound images. The relationship between MAFLD and nephrolithiasis was analyzed using a multivariate logistic regression model and subgroup analysis was performed. Results The proportion of participants with MAFLD was significantly higher in the nephrolithiasis group compared to the non-nephrolithiasis group (47.70% vs. 30.45%, P < 0.001). Multivariate logistic regression analysis showed a significant positive association between MAFLD and nephrolithiasis (adjusted OR=1.38, 95% CI: 1.29 to 1.47). Subgroup analyses indicated that, even after accounting for various factors such as age, diabetes, hypertension, obesity, lipid profiles, and renal function, the positive association between MAFLD and an increased risk of nephrolithiasis remained consistent. Further subgroup analysis revealed that in male patients with MAFLD, the risk of nephrolithiasis increased progressively with increasing severity of liver steatosis. The adjusted multivariable odds ratios were 1.43 (95% CI: 1.33 to 1.53) for mild, 1.48 (95% CI: 1.32 to 1.67) for moderate, and 1.94 (95% CI: 1.47 to 2.58) for severe hepatic steatosis. Conclusions This study found a significant positive association between MAFLD and nephrolithiasis. The risk of nephrolithiasis in males with MAFLD increased substantially with increasing severity of liver steatosis. Therefore, it is essential to strengthen prevention and screening for nephrolithiasis in individuals with MAFLD. More research is needed to elucidate the physiological and pathological mechanisms between MAFLD and nephrolithiasis.
Collapse
Affiliation(s)
- Shengqi Zheng
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Tianchi Hua
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guicao Yin
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Wei Zhang
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoxiang Wang
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Lezhong Qi
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiayong Jing
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qibing Fan
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoping Yu
- Department of Health Promotion Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yifan Li
- Department of Urology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
32
|
Tawfik MY, Amer SAAM, Fouad AM. Shiftwork and insulin resistance in professional drivers: exploring the association using non-insulin-based surrogate measures. BMC Public Health 2025; 25:191. [PMID: 39819581 PMCID: PMC11740691 DOI: 10.1186/s12889-024-21243-9] [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: 05/18/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Previous research has made use of the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) index to explore the association between shiftwork (SW) and insulin resistance (IR). However, the limitations of the HOMA-IR index restrict its use. This study aimed to investigate the relationship between SW and IR in professional drivers using four alternative non-insulin-based IR surrogate measures (NIRS), and to determine the predictors of elevated NIRS. METHODS A comparative cross-sectional study was conducted on professional drivers at four Egyptian companies, where 187 SW were compared to 193 dayworkers (DW). Measurements included: sociodemographic, work, and clinical characteristics. Laboratory and NIRS data included: triglyceride glucose (TyG), triglyceride glucose-body mass index (TyG-BMI), triglyceride to high density lipoprotein cholesterol (TG/HDL-C), and metabolic score of insulin resistance (METS-IR). Further assessments included insomnia severity index (ISI), and perceived stress scale (PSS-10). RESULTS Shiftwork-drivers showed significantly higher levels of NIRS compared to DW-drivers. Shiftwork was significantly associated with elevated TyG (OR: 5.04, 95% CI: 1.98-12.84), TyG-BMI (OR: 4.50, 95% CI: 2.45-8.26), and METS-IR (OR: 6.30, 95% CI: 2.72-14.58). Significant interactions between SW and insomnia or meal-timing habits existed, where SW-drivers with clinically significant insomnia had 11 times higher odds of elevated TyG compared to DW drivers without insomnia. Likewise, SW-drivers experiencing poor meal timing habits had 5.5- and 6.8-times higher odds of elevated TG/HDL-C and METS-IR, respectively, compared to DW divers without poor meal timing habits. Other significant predictors for elevated NIRS included: age, income, stress, overweight/obesity, and poor meal timing habits. CONCLUSIONS This study demonstrates a significant association between shiftwork and elevated insulin resistance in professional drivers. Insomnia and poor meal timing habits significantly increases the odds of insulin resistance among professional drivers, suggesting interventions targeting sleep quality, meal timing, and stress management.
Collapse
Affiliation(s)
- Mirella Youssef Tawfik
- Department of Public health, Occupational and Environmental Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Shaimaa A A M Amer
- Department of Public health, Occupational and Environmental Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ahmed Mahmoud Fouad
- Department of Public health, Occupational and Environmental Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
33
|
Zakaria SS, Hanafy SM. Unraveling the Beneficial Role of Resveratrol in Fructose-Induced Non-Alcoholic Steatohepatitis with a Focus on the AMPK/Nrf2 Signaling Axis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:139. [PMID: 39859121 PMCID: PMC11767180 DOI: 10.3390/medicina61010139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/03/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Background and Objectives: High fructose intake is associated with non-alcoholic fatty liver disease (NAFLD), a chronic liver disease that is on the rise worldwide. New alternatives for treatment, such as bioactive phytochemicals, are needed. The aim of this study was to investigate the beneficial role of resveratrol in treating non-alcoholic steatohepatitis (NASH). Materials and Methods: Sixty male albino rats were allocated to three groups: group I, the normal control group; group II, the fructose-enriched diet group (FED), which was fed a 70% fructose diet for six weeks to induce NASH; and group III, the resveratrol-FED group (RES + FED), which was given the same FED diet plus an oral dose of 70 mg/kg resveratrol (RES) every day for an additional six weeks. We performed histological evaluations and assessed blood lipids and liver enzymes to study resveratrol's impact on NASH. Quantitative real-time PCR was used to assess the mRNA expression of nuclear factor E2-related factor 2 (Nrf2) in the liver samples. ELISA was used to measure Beclin 1, AMPK, IL-6, and the DNA-binding activity of Nrf2. Oxidative stress indicators, including GSH, SOD, and MDA, were evaluated spectrophotometrically. Results: Resveratrol effectively alleviated the biochemical and histopathological abnormalities associated with NASH, improving autophagy by raising Beclin 1 levels while reducing inflammation by decreasing IL-6 levels. Furthermore, resveratrol restored the liver architecture and the oxidative balance, as evidenced by the decreased MDA levels and improved antioxidant status via elevated GSH and SOD activities, as well as the activation of the AMPK/Nrf2 signaling axis. Conclusions: This study specifically examines resveratrol's therapeutic effects in a high-fructose diet-induced NASH model, focusing on the AMPK/Nrf2 signaling pathway to address oxidative stress and autophagy, providing novel insights into its molecular mechanism of action. Resveratrol reduces NASH by boosting autophagy and activating the AMPK/Nrf2 pathway. These findings underscore the potential of resveratrol as a promising therapeutic agent that can support treatment alongside conventional medications in the management of non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- Soha S. Zakaria
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia
| | - Safaa M. Hanafy
- Department of Anatomy and Physiology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13317, Saudi Arabia;
| |
Collapse
|
34
|
Guo J, Mutailipu K, Wen X, Yin J, You H, Qu S, Chen H, Bu L. Association between lymphocyte to high-density lipoprotein cholesterol ratio and insulin resistance and metabolic syndrome in US adults: results from NHANES 2007-2018. Lipids Health Dis 2025; 24:9. [PMID: 39794792 PMCID: PMC11721163 DOI: 10.1186/s12944-024-02411-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Insulin resistance (IR) and metabolic syndrome (MetS) are significant global health challenges that increase the risk of various chronic diseases. The lymphocyte-to-high-density lipoprotein cholesterol ratio (LHR) has emerged as a novel inflammatory metabolic marker. The present study focused on evaluating the association between the LHR and both IR and MetS. METHODS We analyzed data from 14,779 adults aged ≥ 20 years from the National Health and Nutrition Examination Survey (2007-2018). To investigate the relationship between LHR and both IR and MetS, we conducted multivariable logistic regression analyses. The reliability of the results was validated through both stratified and sensitivity analyses. Furthermore, we thoroughly examined possible nonlinear associations by implementing a restricted cubic spline in conjunction with a threshold effect analysis. RESULTS Compared to the lowest LHR quartile, individuals in the highest quartile indicated significantly increased prevalence of IR (odds ratio = 3.72, 95% confidence intervals: 3.01-4.59) and MetS (odds ratio = 11.38, 95% confidence intervals: 8.85-14.63) in fully adjusted models. Subgroup analyses demonstrated that the association between the LHR and IR remained consistent across all subgroups, with no significant interaction effect observed. However, the association between LHR and MetS was more pronounced in female participants. Restricted cubic spline analyses revealed nonlinear associations between LHR and both IR and MetS. The threshold effect analyses identified inflection points at 0.055 for these non-linear relationships. CONCLUSIONS An elevated LHR was positively associated with the prevalence of IR and MetS, indicating its promising role in early screening and disease prevention through biological monitoring.
Collapse
Affiliation(s)
- Junwei Guo
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Kelibinuer Mutailipu
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Xin Wen
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Jiajing Yin
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Hui You
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Shen Qu
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Haibing Chen
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China
| | - Le Bu
- Institute of Obesity, Institute of Thyroid Diseases, Shanghai Center of Thyroid Diseases, Department of Endocrinology and Metabolism, Division of Metabolic Surgery for Obesity and Diabetes, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
35
|
Armisen J, Rauschecker M, Sarv J, Liljeblad M, Wernevik L, Niazi M, Knöchel J, Eklund O, Sandell T, Sherwood J, Bergenholm L, Hallén S, Wang S, Kamble P, Bhat M, Maxvall I, Wang Y, Lee RG, Bhanot S, Guo S, Romeo S, Lawitz E, Fjellström O, Lindén D, Blau JE, Loomba R. AZD2693, a PNPLA3 antisense oligonucleotide, for the treatment of MASH in 148M homozygous participants: Two randomized phase I trials. J Hepatol 2025:S0168-8278(25)00003-0. [PMID: 39798707 DOI: 10.1016/j.jhep.2024.12.046] [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: 08/20/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS A common genetic variant (rs738409) encoding an isoleucine to methionine substitution at position 148 in the PNPLA3 protein is a determinant of hepatic steatosis, inflammation, fibrosis, cirrhosis, and liver-related mortality. AZD2693 is a liver-targeted antisense oligonucleotide against PNPLA3 mRNA. We evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics in single and multiple ascending dose studies. METHODS AZD2693 was assessed in 3D cultures of homozygous PNPLA3 148M primary human hepatocytes and mice expressing human PNPLA3. The single ascending dose study investigated 2-110 mg doses in overweight/mildly obese but otherwise healthy volunteers. The multiple ascending dose study investigated three monthly doses (25 mg, 50 mg and 80 mg) in participants with MRI-proton density fat fraction (MRI-PDFF) ≥7%. Changes in liver fat content were assessed at baseline, weeks 8 and 12 by MRI-PDFF. PNPLA3 mRNA and protein knockdown levels were evaluated for the 80 mg dose. RESULTS AZD2693 potently reduced PNPLA3 expression in human hepatocytes and livers of mice. Clinically, AZD2693 was generally well tolerated (no adverse events leading to discontinuation or treatment-related serious adverse events). Half-life was 14-33 days across investigated doses. A least-square mean liver PNPLA3 mRNA knockdown of 89% and reduction of protein levels demonstrated target engagement. Changes in hepatic steatosis at week 12 were -7.6% and -12.2% (placebo-corrected least-square means) for the 25 mg and 50 mg doses, respectively. There was a dose-dependent increase of polyunsaturated fatty acids in serum triglycerides and decreases vs. placebo in high-sensitivity C-reactive protein and interleukin 6. CONCLUSIONS AZD2693 reduced liver PNPLA3 with an acceptable safety and tolerability profile. These findings support the continued development of AZD2693. IMPACT AND IMPLICATIONS Clinical treatment options for metabolic dysfunction-associated steatohepatitis (MASH) are limited. The genetic risk factor with the largest effect size for progressing to poor liver-related outcomes in MASH is a single-nucleotide polymorphism in the gene PNPLA3 (p.I148M). In phase I single and multiple ascending dose studies, AZD2693, a liver-targeted antisense oligonucleotide, was well tolerated, reduced liver PNPLA3 mRNA and protein levels, and dose-dependently reduced liver fat content in homozygous PNPLA3 148M risk allele carriers. These data support continued development of AZD2693 as a potential precision medicine treatment for MASH. The phase IIb FORTUNA study is now ongoing. CLINICAL TRIAL NUMBER NCT04142424, NCT04483947.
Collapse
Affiliation(s)
- Javier Armisen
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Cambridge, UK
| | - Mitra Rauschecker
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Janeli Sarv
- Late Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mathias Liljeblad
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda Wernevik
- Clinical Operations, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Mohammad Niazi
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Jane Knöchel
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Olof Eklund
- Global Patient Safety, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Therése Sandell
- Global Patient Safety, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - James Sherwood
- Precision Medicine and Biosamples, Diagnostic and HBS Science, Biopharma Diagnostics, Oncology, R&D, AstraZeneca, Cambridge, UK
| | - Linnéa Bergenholm
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefan Hallén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Shan Wang
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Prasad Kamble
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maria Bhat
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Ingela Maxvall
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gothenburg, Sweden
| | - Yixin Wang
- Image Analysis & Platform, Pathology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | | | - Shuling Guo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Eric Lawitz
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ola Fjellström
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Daniel Lindén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jenny E Blau
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals, R&D, AstraZeneca, Gaithersburg, MD, USA.
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, La Jolla, CA, USA
| |
Collapse
|
36
|
Park SH, Park J, Kim H, Lee J, Kwon SY, Lee YB, Kim G, Jin SM, Hur KY, Kim JH. The association of fatty liver index and metabolic syndrome with cardiovascular outcomes, liver-related mortality, and all-cause mortality: a nationwide cohort study. Intern Emerg Med 2025; 20:105-117. [PMID: 39235708 DOI: 10.1007/s11739-024-03758-6] [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: 05/23/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
We investigated the risk of cardiovascular events, all-cause mortality, and liver-related mortality according to the presence of metabolic syndrome (MetS) and fatty liver index (FLI). In this retrospective longitudinal population-based cohort study, we used Korean National Health Insurance Service data from 2009 to 2012. Nonalcoholic fatty liver disease (NAFLD) was defined as FLI ≥ 60. Risk of all-cause mortality, liver-related mortality, and major adverse cardiovascular events (MACE) including myocardial infarction (MI), stroke, heart failure (HF), and cardiovascular disease (CVD)-related mortality was assessed according to the presence of MetS and FLI among adults (aged 40 to 80 years) who underwent health examinations (n = 769,422). During a median 8.59 years of follow up, 44,356 (5.8%) cases of MACE, 24,429 (3.2%) cases of all-cause mortality, and 1114 (0.1%) cases of liver-related mortality were detected in the entire cohort. When the FLI < 30 without MetS group was set as a reference, the FLI ≥ 60 with MetS group had the highest risk of MACE (adjusted hazard ratio [aHR] 2.05, 95% confidence interval [CI] 1.98-2.13) and all-cause mortality (aHR 1.96, 95% CI 1.86-2.07). The risk of liver-related mortality (aHR 10.71, 95% CI 8.05-14.25) was highest in the FLI ≥ 60 without MetS group. The FLI ≥ 60 with MetS group had a higher risk of MACE (aHR 1.39, 95%CI 1.28-1.51), a lower risk of liver-related mortality (aHR 0.44, 95%CI 0.33-0.59), and no significant difference in all-cause mortality compared with the FLI ≥ 60 without MetS group. The FLI ≥ 60 with MetS group was associated with the highest risk of MACE and the FLI ≥ 60 without MetS group had the highest risk liver-related mortality, but there was no significant difference in all-cause mortality between two groups. In conclusion, as FLI levels increase, the risk of MACE increases, and the risk increases additively in the presence of MetS. The risk of liver-related mortality increases with higher FLI levels, the effect of high FLI on increased risk is more significant in groups without MetS compared to those with MetS.
Collapse
Affiliation(s)
- So Hee Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Inje University Ilsan Paik Hospital, College of Medicine, Inje University, Goyang, Republic of Korea
| | - Jiyun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, Republic of Korea
| | - Hasung Kim
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Republic of Korea
| | - Jungkuk Lee
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Republic of Korea
| | - So Yoon Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Daegu Catholic University Hospital, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
| | - You-Bin Lee
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Sang-Man Jin
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Kyu Yeon Hur
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
37
|
Wu Z, Ren M, Tan M, Yang B, Chen S, Yang F, Yuan G, Tan J. Identification and Validation of T Cell-Related Hub Biomarkers for Early Diagnosis of Diabetic Kidney Disease Using Single-Cell and Bulk Dataset Analysis. Crit Rev Eukaryot Gene Expr 2025; 35:65-84. [PMID: 40228227 DOI: 10.1615/critreveukaryotgeneexpr.2025056960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Diabetic kidney disease (DKD) is the most common complication of diabetes and a leading cause of chronic kidney disease that frequently leads to end-stage renal disease (ESRD). The pathogenesis of DKD is complex and is not fully understood. This study was designed to identify key targets for DKD diagnosis and explore the underlying molecular mechanisms. METHODS DKD-specific clusters were selected from single-cell datasets. Gene modules were identified using hairpin-dynamic weighted gene co-expression network analysis (hdWGCNA). Multiple machine learning algorithms were applied to model and screen hub genes from two bulk datasets. Rat model of DKD was built using optical microscopes to observe the histopathological changes in the kidney by HE, PAS, and Masson staining. The expression of RASGRP3, PDE3B, and CD247 in DKD-Rat was verified by RT-PCR, and the expression of RASGRP3, PDE3B, and CD247 in the serum samples of DKD patients was verified by ELISA. The results of sex and age, RASGRP3, PDE3B, CD247 were calculated by multivariate logistic regression analysis. RESULTS Three hub genes were obtained through screening single-cell and two bulk datasets. In-depth exploration of the potential molecular mechanisms of the hub genes was conducted using gene set variation analysis (GSVA), immune infiltration analysis, and single-cell correlation analysis. Receiver operating characteristic (ROC) curve confirmed a high diagnostic value of the hub biomarkers, and a high-efficiency diagnostic model was constructed and mutually validated in the two datasets. We found that damaged tubular number and interstitial fibrotic percentage were significantly increased in DKD rat. As shown by HE, PAS and Masson staining, the mRNA levels of PDE3B and CD247 were markedly upregulated in DKD rat compared with those in the control group. Lower expression levels of RASGRP3 mRNA were manifested in DKD. The levels of RASGRP3, PDE3B, CD247 in DKD patients by ELISA were statistically significant (p < 0.05). PDE3B and CD247 had an AUC value greater than 0.9,RASGRP3 had an AUC value greater than 0.7. CONCLUSION This study identified 3 T cell-related hub biomarkers, providing references for the early diagnosis of DKD and changes in T cells during DKD progression.
Collapse
Affiliation(s)
| | - Meifang Ren
- Department of Nephrology, The First Affiliated Hospital of Hebei University of Chinese Medicine, 050000, China
| | - Miao Tan
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, 050011, China
| | - Bing Yang
- Graduate School, Hebei University of Chinese Medicine, 050091, China
| | - Suzhi Chen
- Department of Nephrology, The First Affiliated Hospital of Hebei University of Chinese Medicine, 050000, China
| | - Fengwen Yang
- Department of Nephrology, The First Affiliated Hospital of Hebei University of Chinese Medicine, 050000, China
| | - Guodong Yuan
- Department of Nephrology, The First Affiliated Hospital of Hebei University of Chinese Medicine, 050000, China
| | - Jinchuan Tan
- Department of Nephrology, The First Affiliated Hospital of Hebei University of Chinese Medicine, 050000, China
| |
Collapse
|
38
|
Zhang K, Yang C, Zhao X, Wang Y, Gu Z, Yang R, Ding H, Li S, Qin J, Chu X. Associations of Urinary Nickel with NAFLD and Liver Fibrosis in the USA: A Nationwide Cross‑Sectional Study. Biol Trace Elem Res 2025; 203:30-38. [PMID: 38514508 DOI: 10.1007/s12011-024-04151-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Despite the robust correlation between metabolic disorders and heavy metals, there has been limited research on the associations between nickel levels and non-alcoholic fatty liver disease (NAFLD) as well as liver fibrosis. This study aimed to examine the associations among urinary nickel, NAFLD, and liver fibrosis. The data utilized in this study were obtained from the National Health and Nutrition Examination Survey 2017-2020. A comprehensive screening process was conducted, resulting in the inclusion of a total of 3169 American adults in the analysis. The measurement of urinary nickel was conducted through inductively coupled-plasma mass spectrometry. Vibration-controlled transient elastography was employed to assess the controlled attenuation parameter and liver stiffness measurement as indicators for NAFLD and liver fibrosis, respectively. Multivariable logistic regression models were employed to evaluate the associations among urinary nickel, NAFLD, and liver fibrosis. Restricted cubic splines were employed to explored the nonlinear associations. After adjusting for all covariates, the correlation between the highest quartile of urinary nickel and NAFLD was found to be significant (OR = 1.65; 95% CI, 1.19-2.27). Subgroup analysis revealed that the correlation was significant only in men. A significant association occurred between the second quartile of urinary nickel and liver fibrosis (OR 1.88; 95% CI, 1.22-2.90). Restricted cubic spline showed that the relationship was linear between urinary nickel and NAFLD and non-monotonic, inverse U-shaped between urinary nickel and liver fibrosis. This cross-sectional study indicated that the risk of NAFLD is associated with urinary nickel, and this correlation was only present among males.
Collapse
Affiliation(s)
- Kening Zhang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Chunxiao Yang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Xue Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Yuanyuan Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Zhuo Gu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Ruiming Yang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Haiyan Ding
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Shuangshuang Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Jian Qin
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China
| | - Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, No.157 Baojian Road, Nangang District, Heilongjiang Province, Harbin, 150081, China.
- Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
39
|
Hobeika C, Ronot M, Guiu B, Ferraioli G, Iijima H, Tada T, Lee DH, Kuroda H, Lee YH, Lee JM, Kim SY, Cassinotto C, Maiocchi L, Raimondi A, Nishimura T, Kumada T, Kwon EY, Jang JK, Correas JM, Valla D, Vilgrain V, Dioguardi Burgio M. Ultrasound-based steatosis grading system using 2D-attenuation imaging: An individual patient data meta-analysis with external validation. Hepatology 2025; 81:212-227. [PMID: 38652643 DOI: 10.1097/hep.0000000000000895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/07/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND AND AIMS Noninvasive tools assessing steatosis, such as ultrasonography-based 2D-attenuation imaging (ATI), are needed to tackle the worldwide burden of steatotic liver disease. This one-stage individual patient data (IPD) meta-analysis aimed to create an ATI-based steatosis grading system. APPROACH AND RESULTS A systematic review (EMBASE + MEDLINE, 2018-2022) identified studies, including patients with histologically or magnetic resonance imaging proton-density fat fraction (MRI-PDFF)-verified ATI for grading steatosis (S0 to S3). One-stage IPD meta-analyses were conducted using generalized mixed models with a random study-specific intercept. Created ATI-based steatosis grading system (aS0 to aS3) was externally validated on a prospective cohort of patients with type 2 diabetes and metabolic dysfunction-associated steatotic liver disease (n=174, histologically and MRI-PDFF-verified steatosis). Eleven enrolled studies included 1374 patients, classified into S0, S1, S2, and S3 in 45.4%, 35.0%, 9.3%, and 10.3% of the cases. ATI was correlated with histological steatosis ( r = 0.60; 95% CI: 0.52, 0.67; p < 0.001) and MRI-PDFF ( r = 0.70; 95% CI: 0.66, 0.73; p < 0.001) but not with liver stiffness ( r = 0.03; 95% CI: -0.04, 0.11, p = 0.343). Steatosis grade was an independent factor associated with ATI (coefficient: 0.24; 95% CI: [0.22, 0.26]; p < 0.001). ATI marginal means within S0, S1, S2, and S3 subpopulations were 0.59 (95% CI: [0.58, 0.61]), 0.69 (95% CI [0.67, 0.71]), 0.78 (95% CI: [0.76, 0.81]), and 0.85 (95% CI: [0.83, 0.88]) dB/cm/MHz; all contrasts between grades were significant ( p < 0.0001). Three ATI thresholds were calibrated to create a new ATI-based steatosis grading system (aS0 to aS3, cutoffs: 0.66, 0.73, and 0.81 dB/cm/MHz). Its external validation showed Obuchowski measures of 0.84 ± 0.02 and 0.82 ± 0.02 with histologically based and MRI-PDFF-based references. CONCLUSIONS ATI is a reliable, noninvasive marker of steatosis. This validated ATI-based steatosis grading system could be valuable in assessing patients with metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Christian Hobeika
- Department of HPB Surgery and Liver Transplantation, AP-HP, Hôpital Beaujon, Clichy, France
- Université Paris Cité, Inserm, CArcinose Péritoine Paris-Technologies, Paris, France
- Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Maxime Ronot
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
| | - Boris Guiu
- Department of Radiology, St-Eloi University Hospital, Montpellier, France
| | - Giovanna Ferraioli
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche, University of Pavia, Pavia, Italy
| | - Hiroko Iijima
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hyogo Medical University, Hyogo, Japan
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Society Himeji Hospital, Hyogo, Japan
| | - Dong Ho Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hidekatsu Kuroda
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Iwate Medical University, Iwate, Japan
| | - Young Hwan Lee
- Department of Radiology, Wonkwang University School of Medicine and Hospital, Iksan, Korea
| | - Jeong Min Lee
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - So Yeon Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | - Laura Maiocchi
- Ultrasound Unit, Dipartimento Servizi Diagnostici e per Immagini Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Ambra Raimondi
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche, University of Pavia, Pavia, Italy
- Ultrasound Unit, Dipartimento Servizi Diagnostici e per Immagini Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Takashi Nishimura
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hyogo Medical University, Hyogo, Japan
| | - Takashi Kumada
- Department of Nursing, Gifu Kyoritsu University, Gifu, Japan
| | - Eun Young Kwon
- Department of Radiology, Wonkwang University School of Medicine and Hospital, Iksan, Korea
| | - Jong Keon Jang
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jean-Michel Correas
- AP-HP, Hôpital Necker Enfants Malades, Service d'Imagerie Adulte, Paris, France
- Sorbonne Université, CNRS, INSERM Laboratoire d'Imagerie Biomédicale, Paris, France
| | - Dominique Valla
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
- Service d'hépatologie, Hôpital Beaujon, Clichy, France
| | - Valérie Vilgrain
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
| | - Marco Dioguardi Burgio
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
| |
Collapse
|
40
|
Goldberg DT, Yaskolka Meir A, Tsaban G, Rinott E, Kaplan A, Zelicha H, Klöting N, Ceglarek U, Iserman B, Shelef I, Rosen P, Blüher M, Stumvoll M, Etzion O, Stampfer MJ, Hu FB, Shai I. Novel proteomic signatures may indicate MRI-assessed intrahepatic fat state and changes: The DIRECT PLUS clinical trial. Hepatology 2025; 81:198-211. [PMID: 38537153 PMCID: PMC11643130 DOI: 10.1097/hep.0000000000000867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/03/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND AND AIMS We demonstrated in the randomized 18-month DIRECT PLUS trial (n = 294) that a Mediterranean (MED) diet, supplemented with polyphenol-rich Mankai duckweed, green tea, and walnuts and restricted in red/processed meat, caused substantial intrahepatic fat (IHF%) loss compared with 2 other healthy diets, reducing NAFLD by half, regardless of similar weight loss. Here, we investigated the baseline proteomic profile associated with IHF% and the changes in proteomics associated with IHF% changes induced by lifestyle intervention. APPROACH AND RESULTS We calculated IHF% by proton magnetic resonance spectroscopy (normal IHF% <5% and abnormal IHF% ≥5%). We assayed baseline and 18-month samples for 95 proteomic biomarkers.Participants (age = 51.3 ± 10.8 y; 89% men; and body mass index = 31.3 ± 3.9 kg/m 2 ) had an 89.8% 18-month retention rate; 83% had eligible follow-up proteomics measurements, and 78% had follow-up proton magnetic resonance spectroscopy. At baseline, 39 candidate proteins were significantly associated with IHF% (false discovery rate <0.05), mostly related to immune function pathways (eg, hydroxyacid oxidase 1). An IHF% prediction based on the DIRECT PLUS by combined model ( R2 = 0.47, root mean square error = 1.05) successfully predicted IHF% ( R2 = 0.53) during testing and was stronger than separately inputting proteins/traditional markers ( R2 = 0.43/0.44). The 18-month lifestyle intervention induced changes in 18 of the 39 candidate proteins, which were significantly associated with IHF% change, with proteins related to metabolism, extracellular matrix remodeling, and immune function pathways. Thrombospondin-2 protein change was higher in the green-MED compared to the MED group, beyond weight and IHF% loss ( p = 0.01). Protein principal component analysis revealed differences in the third principal component time distinct interactions across abnormal/normal IHF% trajectory combinations; p < 0.05 for all). CONCLUSIONS Our findings suggest novel proteomic signatures that may indicate MRI-assessed IHF state and changes during lifestyle intervention. Specifically, carbonic anhydrase 5A, hydroxyacid oxidase 1, and thrombospondin-2 protein changes are independently associated with IHF% change, and thrombospondin-2 protein change is greater in the green-MED/high polyphenols diet.
Collapse
Affiliation(s)
- Dana T. Goldberg
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Yaskolka Meir
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gal Tsaban
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ehud Rinott
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Kaplan
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hila Zelicha
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nora Klöting
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Berend Iserman
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry, and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Ilan Shelef
- Department of Diagnostic Imaging, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Philip Rosen
- Department of Diagnostic Imaging, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Ohad Etzion
- Department of Gastroenterology and Liver Diseases, Soroka University Medical Center, Beersheba, Israel
| | - Meir J. Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B. Hu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Iris Shai
- The Health & Nutrition Innovative International Research Center, Department of Epidemiology, Biostatistics and Community Health Sciences, Faculty of Health Sciences, School of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Medicine, University of Leipzig, Leipzig, Germany
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
41
|
Nelson LW, Lee MH, Garrett JW, Pickhardt SG, Warner JD, Summers RM, Pickhardt PJ. Intrapatient Changes in CT-Based Body Composition After Initiation of Semaglutide (Glucagon-Like Peptide-1 Receptor Agonist) Therapy. AJR Am J Roentgenol 2024:1-10. [PMID: 39230989 DOI: 10.2214/ajr.24.31805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
BACKGROUND. The long-acting glucagon-like peptide-1 receptor agonist semaglutide is used to treat type 2 diabetes or obesity in adults. Clinical trials have observed associations of semaglutide with weight loss, improved control of diabetes, and cardiovascular risk reduction. OBJECTIVE. The purpose of this study was to evaluate intrapatient changes in body composition after initiation of semaglutide therapy by applying an automated suite of CT-based artificial intelligence (AI) body composition tools. METHODS. This retrospective study included adult patients who were receiving semaglutide treatment and who, between January 2016 and November 2023, underwent abdominopelvic CT within both 5 years before and 5 years after initiation of semaglutide. An automated suite of previously validated CT-based AI body composition tools was applied to scans obtained before semaglutide initiation (hereafter, presemaglutide scans) and scans obtained after semaglutide initiation (hereafter, postsemaglutide scans) to quantify visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) area, skeletal muscle area and attenuation, intermuscular adipose tissue (IMAT) area, liver volume and attenuation, and trabecular bone mineral density (BMD). Patients with weight loss of 5 kg or more and those with weight gain of 5 kg or more between the scans were compared. RESULTS. The study included 241 patients (151 women and 90 men; mean age, 60.4 ± 12.4 [SD] years). In the weight-loss group (n = 67), the postsemaglutide scan, compared with the presemaglutide scan, showed a decrease in VAT area (309.4 vs 341.1 cm2, p < .001), SAT area (371.4 vs 410.7 cm2, p < .001), muscle area (179.2 vs 193.0, p < 0.001), and liver volume (2379.0 vs 2578 HU, p = .009) and an increase in liver attenuation (74.5 vs 67.6 HU, p = .03). In the weight-gain group (n = 48), the postsemaglutide scan, compared with the presemaglutide scan, showed an increase in VAT area (334.0 vs 312.8, p = .002), SAT area (485.8 vs 448.8 cm2, p = .01), and IMAT area (48.4 vs 37.6, p = .009) and a decrease in muscle attenuation (5.9 vs 13.1, p < .001). Other comparisons were not statistically significant (p > .05). CONCLUSION. Patients using semaglutide who lost versus gained weight showed distinct patterns of changes in CT-based body composition measures. Those with weight loss had overall favorable shifts in measures related to cardiometabolic risk. A decrease in muscle attenuation in those with weight gain is consistent with decreased muscle quality. CLINICAL IMPACT. Among patients using semaglutide, automated CT-based AI tools provide biomarkers of changes in body composition beyond those that are evident by standard clinical measures.
Collapse
Affiliation(s)
- Leslie W Nelson
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - Matthew H Lee
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - John W Garrett
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Silas G Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - Joshua D Warner
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
| | - Ronald M Summers
- Imaging Biomarkers and Computer-Aided Diagnosis Laboratory, Radiology and Imaging Sciences, NIH Clinical Center, Bethesda, MD
| | - Perry J Pickhardt
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, E3/311 Clinical Science Center, 600 Highland Ave, Madison, WI 53792-3252
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
42
|
Bucci T, Nabrdalik K, Baratta F, Pastori D, Pignatelli P, Hydes T, Alam U, Violi F, Lip GYH. Risk of Adverse Events in Anticoagulated Patients With Atrial Fibrillation and Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2024; 110:208-217. [PMID: 38864452 PMCID: PMC11651694 DOI: 10.1210/clinem/dgae394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/15/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND The clinical impact of nonalcoholic fatty liver disease (NAFLD) in patients with atrial fibrillation (AF) is still controversial. AIM To evaluate the 1-year risk of all-cause death, thromboembolic events, and bleeding in patients with AF-NAFLD. METHODS Retrospective study with a health research network (TriNetX). Patients with AF on oral anticoagulation (OAC) were categorized according to the presence of NAFLD into 2 groups. The primary outcomes were the 1-year risks of (1) a composite cardiovascular outcome (all-cause death, myocardial infarction, stroke, cardiac arrest, and pulmonary embolism) and (2) a composite hemorrhagic outcome (intracranial hemorrhage and gastrointestinal bleeding). Cox regression analysis before and after propensity score matching was used to estimate hazard ratio (HR) and 95% 95% CI,. Sensitivity analyses investigated the risk associated with cirrhosis, thrombocytopenia, and type of OAC (warfarin vs non-vitamin K antagonist oral anticoagulants (NOACs). RESULTS We identified 22 636 patients with AF-NAFLD (69 ± 12 years, 46.7% females) and 391 014 patients with AF and without liver disease (72 ± 12 years, 42.7% females). NAFLD was associated with a higher risk of composite cardiovascular (HR, 1.54; 95% CI, 1.47-1.61) and hemorrhagic (HR, 1.56; 95% CI, 1.42-1.72) outcomes. This was consistent also for all the single outcomes. Cirrhotic and thrombocytopenic patients with AF-NAFLD showed the highest risks. Compared to patients with AF-NAFLD on NOACs, those on warfarin were associated with a higher risk of cardiovascular and hemorrhagic outcomes. CONCLUSION In patients with AF, NAFLD is associated with a higher 1-year risk of adverse events, with the risk of adverse events progressively increasing from noncirrhotic to cirrhotic and from nonthrombocytopenic to thrombocytopenic patients. NOACs were associated with a better effectiveness and safety profile compared to warfarin.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Katarzyna Nabrdalik
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice 40-055, Poland
| | - Francesco Baratta
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Theresa Hydes
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Diabetes & Endocrinology Research and Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool and Liverpool University Hospital NHS Foundation Trust, Liverpool L69 7ZX, UK
| | - Uazman Alam
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Diabetes & Endocrinology Research and Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool and Liverpool University Hospital NHS Foundation Trust, Liverpool L69 7ZX, UK
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg 9000, Denmark
| |
Collapse
|
43
|
You Y, Pei X, Jiang W, Zeng Q, Bai L, Zhou T, Lv X, Tang H, Wu D. Non-obese non-alcoholic fatty liver disease and the risk of chronic kidney disease: a systematic review and meta-analysis. PeerJ 2024; 12:e18459. [PMID: 39713133 PMCID: PMC11660860 DOI: 10.7717/peerj.18459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/14/2024] [Indexed: 12/24/2024] Open
Abstract
Background Data on risk of developing chronic kidney disease (CKD) between non-obese and obese non-alcoholic fatty liver disease (NAFLD) patients are limited. We aimed to reveal the risk difference of incident CKD between non-obese and obese NAFLD patients. Methods We searched PubMed, Embase, and Web of Science databases for studies which reported the incidence of CKD in non-obese and obese NAFLD from inception to 10 March 2024. The primary and secondary outcomes were pooled. Subgroup analysis was used to examine the heterogeneity. Results A total of 15 studies were incorporated. The incidence of CKD in non-obese and obese NAFLD were 1,450/38,720 (3.74%) and 3,067/84,154 (3.64%), respectively. Non-obese NAFLD patients had a comparable risk of CKD as obese NAFLD (odds ratio [OR] 0.92, 95% confidence interval [95% CI] [0.72-1.19], I2 = 88%). No differences in estimated glomerular filtration rate and serum creatinine between non-obese and obese NAFLD were found. The mean differences (MD) and 95% CI were 0.01 [-0.02 to 0.04] and 0.50 [-0.90 to 1.90], respectively. In subgroup analyses, non-obese NAFLD had higher eGFR when diagnosed with ultrasound (MD 1.45, 95% CI [0.11-2.79], I2 = 21%). Non-obese NAFLD had higher creatinine in non-Asian (MD 0.06, 95% CI [0.01-0.11], I2 = 55%) and when taking BMI > 30 as the criterion for obesity (MD 0.06, 95% CI [0.00-0.12], I2 = 76%). The occurrence of CKD did not differ when non-obese NAFLD were categorized into overweight and normal-weight types. Conclusions Non-obese NAFLD patients experienced the same risk of CKD compared to obese NAFLD.
Collapse
Affiliation(s)
- Yixian You
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiong Pei
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Taoyou Zhou
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoju Lv
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Singla B. Fanlian Huazhuo Formula: A promising herbal preparation for metabolic liver disease. World J Gastroenterol 2024; 30:4964-4968. [PMID: 39679304 PMCID: PMC11612710 DOI: 10.3748/wjg.v30.i46.4964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has increased significantly in recent decades and is projected to increase further due to the rising obesity rates. MASLD patients are at higher risk of developing advanced liver diseases "cirrhosis and hepatocellular carcinoma" as well as liver- or cardiovascular-related mortality. Existing lipid-lowering therapies failed to reduce the risk of mortality in these patients. Therefore, there is an urgent need for pharmacotherapies that can control and even reverse this disease. Fanlian Huazhuo Formula (FLHZF) is a combination herbal preparation, and its various individual constituents regulate hepatic lipid metabolism, adipose tissue inflammation, and gut microbiota. Despite, these useful effects, limited information is available on its benefits in diet-induced hepatosteatosis. In this article, we discuss the research findings recently published about the therapeutic effects of FLHZF in suppressing MASLD development and underlying mechanisms. Utilizing a series of in vitro and in vivo experiments, the authors demonstrated for the first time that FLHZF suppresses MASLD in male mice possibly by inhibiting hepatic de novo lipogenesis pathways and reducing hepatocyte death. This study paves the way for future investigations aimed at investigating FLHZF's role in inhibiting lipogenesis particularly using radioactively-labeled glucose and acetate, and governing hepatocyte mitochondrial function, gut microbiome profile, and its effects in other models of MASLD, and female mice.
Collapse
Affiliation(s)
- Bhupesh Singla
- Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38103, United States
| |
Collapse
|
45
|
Xie K, Chen M, An H, Gao J, Tang C, Huang Z. Causal associations of immunophenotypes with metabolic dysfunction-associated fatty liver disease and mediating pathways: a Mendelian randomization study. Ther Adv Chronic Dis 2024; 15:20406223241303649. [PMID: 39669435 PMCID: PMC11635899 DOI: 10.1177/20406223241303649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024] Open
Abstract
Background Increasing evidence suggests that immunophenotypes play a crucial role in Metabolic dysfunction-associated fatty liver disease (MAFLD), but the specific immunophenotypes contributing to its pathogenesis remain unclear. Objectives This study aimed to elucidate the causal associations between immunophenotypes and MAFLD and identify the underlying mediation pathways involved. Design Mendelian randomization (MR) study. Methods This study is a quasi-causal inference analysis using univariable and multivariable MR (UVMR and MVMR). Five MAFLD genome-wide association studies (GWASs) and the largest immunophenotype GWAS were analyzed to assess their causal associations. Two-step MR identified potential mediators and quantified their mediation proportions. Comprehensive MR methods, multiple sensitivity analyses, meta-analyses, and false discovery rate (FDR) further enhanced the robustness of our findings. Results Pooled inverse-variance weighted (IVW) estimates in UVMR identified 47 immunophenotypes having a suggestive causal association with MAFLD. After adjusting for FDR, three lymphocyte phenotypes remained significant: CD20 on IgD-CD24- B cells (OR: 1.035, p fdr: 0.006), terminally differentiated CD8+ T cells %T cells (OR: 1.052, p fdr: 0.006), and CD4 on CD39+ secreting CD4+ regulatory T cells (OR: 1.036, p fdr: 0.046). Meta-analysis of IVW MVMR estimates with confounders adjustment confirmed that CD20 on IgD-CD24- B cells and terminally differentiated CD8+ T cells %T cells had significant direct causal associations on MAFLD (p fdr < 0.05). Additionally, two-step MR analysis identified the waist-to-hip ratio as a mediator, accounting for 42.64% of the causal association between CD20 on IgD-CD24- B cells and MAFLD. Conclusion The causal associations of three lymphocyte phenotypes with increased MAFLD risk were identified in this study. CD20 on IgD-CD24- B cells may both directly and indirectly elevate MAFLD risk, while terminally differentiated CD8+ T cells have a direct causal relationship with MAFLD. These findings suggest new possibilities for targeted therapies and underscore the potential for personalized immunotherapy in managing MAFLD.
Collapse
Affiliation(s)
- Kexin Xie
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Chen
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongjin An
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyin Huang
- Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No. 37, Guoxue Xiang, Chengdu 610041, China
| |
Collapse
|
46
|
Zhang C, Cao L, Xu B, Zhang W. Interaction between trouble sleeping and diabetes on metabolic dysfunction-associated fatty liver disease and liver fibrosis in adults results from the National Health and Nutrition Examination Survey 2017-2018. Eur J Gastroenterol Hepatol 2024; 36:1437-1446. [PMID: 39373628 PMCID: PMC11527372 DOI: 10.1097/meg.0000000000002860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD), trouble sleeping, and diabetes, as major public health problems, were closely related. The study examined the interaction between trouble sleeping and diabetes on MAFLD and liver fibrosis in adults with MAFLD. METHODS The data were obtained from the National Health and Nutrition Examination Survey 2017-2018. Multivariate logistic regression model and subgroup analyses were conducted to assess the relationship between either trouble sleeping or diabetes on MAFLD and liver fibrosis. Relative excess risk due to interaction (RERI), attributable proportion of interaction (AP), and synergy index (S) were utilized to assess the additive interaction. RESULTS Ultimately, 3747 participants were included, with 2229 known MAFLD subjects. Compared with participants without diabetes, those with diabetes had a higher risk of MAFLD [odds ratio (OR) = 5.55; 95% confidence interval (CI) = 4.07-7.56] and liver fibrosis risk (OR = 3.61; 95% CI = 2.67-4.89). We also found a significant association of trouble sleeping with an increased risk of MAFLD (OR = 1.54; 95% CI = 1.17-2.02) and liver fibrosis risk (OR = 1.51; 95% CI = 1.06-2.16), compared with those without trouble sleeping. Moreover, there was a significant interaction between diabetes and trouble sleeping on MAFLD [RERI = 1.76 (95% CI: -0.22 to 3.73), AP = 0.35 (95% CI: 0.08-0.63), S = 1.80 (95% CI: 1.02-3.16)] and liver fibrosis risk [RERI = 1.79 (95% CI: 0.37-3.21), AP = 0.44 (95% CI: 0.20-0.69), S = 2.44 (95% CI: 1.18-5.08)]. CONCLUSION The findings highlight that trouble sleeping and diabetes had a synergistic effect on MAFLD and liver cirrhosis. The study highlights the importance of addressing both trouble sleeping and diabetes management in adults to mitigate the risk of MAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Cui Zhang
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| | - Lili Cao
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| | - Bo Xu
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| | - Wei Zhang
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
47
|
Mateo-Marín MA, Alves-Bezerra M. Targeting acetyl-CoA carboxylases for the treatment of MASLD. J Lipid Res 2024; 65:100676. [PMID: 39461620 PMCID: PMC11621487 DOI: 10.1016/j.jlr.2024.100676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Hepatic accumulation of triglycerides is a hallmark feature of metabolic dysfunction-associated steatotic liver disease (MASLD). Growing evidence indicates that increased rates of de novo lipogenesis (DNL) are one of the earliest metabolic changes promoting hepatic steatosis in the onset of MASLD. The first step in DNL is catalyzed by acetyl-CoA carboxylases (ACC), which mediate the conversion of acetyl-CoA into malonyl-CoA. Given the critical role of ACC enzymes on DNL, ACC-based therapies have emerged as an attractive approach to address MASLD, leading to the development of pharmacologic inhibitors of ACC. In clinical trials, several of those compounds led to decreased DNL rates and improved hepatic steatosis in patients with MASLD. In this review, we describe the development of ACC dual inhibitors and isoform-specific inhibitors along with their clinical testing using monotherapy and combination therapy approaches. We also discuss their efficacy and safety profiles, identifying potential directions for future research. It is anticipated that advances in ACC-based therapies will be critical to the management of MASLD.
Collapse
Affiliation(s)
- María Antonia Mateo-Marín
- Department of Biomedicine, Biotechnology and Public Health, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Faculty of Medicine, University of Cadiz, Cadiz, Spain
| | - Michele Alves-Bezerra
- Department of Biomedicine, Biotechnology and Public Health, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Faculty of Medicine, University of Cadiz, Cadiz, Spain.
| |
Collapse
|
48
|
Babu AF, Palomurto S, Kärjä V, Käkelä P, Lehtonen M, Hanhineva K, Pihlajamäki J, Männistö V. Metabolic signatures of metabolic dysfunction-associated steatotic liver disease in severely obese patients. Dig Liver Dis 2024; 56:2103-2110. [PMID: 38825414 DOI: 10.1016/j.dld.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024]
Abstract
BACKROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) can lead to liver fibrosis, cirrhosis, and hepatocellular carcinoma. Still, most patients with MASLD die from cardiovascular diseases indicating metabolic alterations related to both liver and cardiovascular pathology. AIMS AND METHODS The aim of this study was to assess biologic pathways behind MASLD progression from steatosis to metabolic dysfunction-associated steatohepatitis (MASH) using non-targeted liquid chromatography-mass spectrometry analysis in 106 severely obese individuals (78 women, mean age 47.7 7 ± 9.2 years, body mass index 41.8 ± 4.3 kg/m²) undergoing laparoscopic Roux-en-Y gastric bypass. RESULTS We identified several metabolites that are associated with MASLD progression. Most importantly, we observed a decrease of lysophosphatidylcholines LPC(18:2), LPC(18:3), and LPC(20:3) and increase of xanthine when comparing those with steatosis to those with MASH. We found that indole propionic acid and threonine were negatively correlated to fibrosis, but not with the metabolic disturbances associated with cardiovascular risk. Xanthine, ketoleucine, and tryptophan were positively correlated to lobular inflammation and ballooning but also with insulin resistance, and dyslipidemia, respectively. The results did not change when taking into account the most important genetic risk factors of MASLD. CONCLUSIONS Our findings suggest that there are several separate biological pathways, some of them independent of insulin resistance and dyslipidemia, associating with MASLD.
Collapse
Affiliation(s)
- Ambrin Farizah Babu
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland
| | - Saana Palomurto
- Department of Surgery, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Pirjo Käkelä
- Department of Surgery, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, Faculty of Health Science, University of Eastern Finland, 70211 Kuopio, Finland; LC-MS Metabolomics Center, Biocenter Kuopio, 70211 Kuopio, Finland
| | - Kati Hanhineva
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, 20014 Turku, Finland
| | - Jussi Pihlajamäki
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70210 Kuopio Finland
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland.
| |
Collapse
|
49
|
Beyoğlu D, Popov YV, Idle JR. Metabolomic Hallmarks of Obesity and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2024; 25:12809. [PMID: 39684520 DOI: 10.3390/ijms252312809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
From a detailed review of 90 experimental and clinical metabolomic investigations of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD), we have developed metabolomic hallmarks for both obesity and MASLD. Obesity studies were conducted in mice, rats, and humans, with consensus biomarker groups in plasma/serum being essential and nonessential amino acids, energy metabolites, gut microbiota metabolites, acylcarnitines and lysophosphatidylcholines (LPC), which formed the basis of the six metabolomic hallmarks of obesity. Additionally, mice and rats shared elevated cholesterol, humans and rats shared elevated fatty acids, and humans and mice shared elevated VLDL/LDL, bile acids and phosphatidylcholines (PC). MASLD metabolomic studies had been performed in mice, rats, hamsters, cows, geese, blunt snout breams, zebrafish, and humans, with the biomarker groups in agreement between experimental and clinical investigations being energy metabolites, essential and nonessential amino acids, fatty acids, and bile acids, which lay the foundation of the five metabolomic hallmarks of MASLD. Furthermore, the experimental group had higher LPC/PC and cholesteryl esters, and the clinical group had elevated acylcarnitines, lysophosphatidylethanolamines/phosphatidylethanolamines (LPE/PE), triglycerides/diglycerides, and gut microbiota metabolites. These metabolomic hallmarks aid in the understanding of the metabolic role played by obesity in MASLD development, inform mechanistic studies into underlying disease pathogenesis, and are critical for new metabolite-inspired therapies.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Yury V Popov
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jeffrey R Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| |
Collapse
|
50
|
Zhou X, Liao J, Liu L, Meng Y, Yang D, Zhang X, Long L. Association of depression with severe non-alcoholic fatty liver disease: evidence from the UK Biobank study and Mendelian randomization analysis. Sci Rep 2024; 14:28561. [PMID: 39557910 PMCID: PMC11574024 DOI: 10.1038/s41598-024-79100-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The relationship between depression and severe non-alcoholic fatty liver disease (NAFLD) has not been clearly defined. We conducted a longitudinal cohort study and a two-sample Mendelian randomization (MR) analysis to assess the association of depression with severe NAFLD risk. We used individual data from the UK Biobank study with 481,181 participants, and summary data from published genome-wide association studies. The association between depression and severe NAFLD was assessed using Cox proportional hazards regression analysis. Two-sample MR for depression with NAFLD was conducted, the principal analysis employed the inverse variance weighted (IVW) approach. In the observational study, after a median follow-up of 13.46 years, 4,563 participants had severe NAFLD. In multivariable-adjusted model, participants with depression had an increased risk of severe NAFLD (hazards ratio:1.21, 95% confidence interval (CI):1.09-1.34), as compared to those without depression. In subgroup analyses, the association between depression and severe NAFLD risk was generally observed across different subgroups. For the MR, result also showed that genetically predicted depression was causally associated with a higher risk of NAFLD (odds ratio:1.55, 95%CI:1.10-2.19) in IVW. Our study revealed a prospective association of depression with severe NAFLD, thus potentially necessitating clinical monitoring of individuals with depression for severe NAFLD.
Collapse
Affiliation(s)
- Xiaorui Zhou
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Juan Liao
- Department of Gastroenterology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Li Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510006, China
| | - Yajing Meng
- Mental Health Center, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dailan Yang
- Department of Gastroenterology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, 02115, USA
| | - Lu Long
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA.
| |
Collapse
|