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
|
Ayares G, Diaz LA, Idalsoaga F, Alkhouri N, Noureddin M, Bataller R, Loomba R, Arab JP, Arrese M. MetALD: New Perspectives on an Old Overlooked Disease. Liver Int 2025; 45:e70017. [PMID: 40179033 PMCID: PMC11967760 DOI: 10.1111/liv.70017] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 04/05/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-associated liver disease (ALD) are the major contributors to the liver disease burden globally. The rise in these conditions is linked to obesity, type 2 diabetes, metabolic syndrome and increased alcohol consumption. MASLD and ALD share risk factors, pathophysiology and histological features but differ in their thresholds for alcohol use, and the ALD definition does not require the presence of metabolic dysfunction. A recent multi-society consensus overhauled the nomenclature of liver steatosis and introduced the term MetALD to describe patients with metabolic dysfunction who drink more than those with MASLD and less than those with ALD. This new terminology aims to enhance the understanding and management of liver disease but poses challenges, such as the need to accurately measure alcohol consumption in research and clinical practice settings. Recent studies show that MetALD has significant implications for patient management, as it is associated with increased mortality risks and more severe liver outcomes compared to MASLD alone. MetALD patients face increased risks of liver disease progression, cancer and cardiovascular disease. The diagnosis of MetALD involves the adequate quantification of alcohol use through standardised questionnaires and/or biomarkers as well as proper assessment of liver disease stage and progression risk using non-invasive tools including serologic markers, imaging, elastography techniques and genetic testing. Effective management requires addressing both metabolic and alcohol-related factors to improve outcomes. This review intends to provide a comprehensive overview of MetALD, covering pathogenesis, potential diagnostic approaches, management strategies and emerging therapies.
Collapse
Affiliation(s)
- Gustavo Ayares
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- Escuela de Medicina, Universidad Finis TerraeSantiagoChile
| | - Luis Antonio Diaz
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- MASLD Research Center, Division of Gastroenterology and HepatologyUniversity of California San DiegoCaliforniaUSA
| | - Francisco Idalsoaga
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- Division of Gastroenterology Department of MedicineSchulich School of Medicine, Western University & London Health Sciences CentreLondonOntarioCanada
| | - Naim Alkhouri
- Department of HepatologyArizona Liver HealthChandlerArizonaUSA
| | | | - Ramon Bataller
- Liver UnitHospital Clinic and Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and HepatologyUniversity of California San DiegoCaliforniaUSA
| | - Juan Pablo Arab
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal MedicineVirginia Commonwealth University School of MedicineVirginiaUSA
| | - Marco Arrese
- Departamento de GastroenterologíaEscuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
3
|
Vianna P, Mehrbod P, Chaudhary M, Eickenberg M, Wolf G, Belilovsky E, Tang A, Cloutier G. Unsupervised Test-Time Adaptation for Hepatic Steatosis Grading Using Ultrasound B-Mode Images. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2025; 72:601-611. [PMID: 40138246 DOI: 10.1109/tuffc.2025.3555180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Ultrasound (US) is considered a key modality for the clinical assessment of hepatic steatosis (i.e., fatty liver) due to its noninvasiveness and availability. Deep learning methods have attracted considerable interest in this field, as they are capable of learning patterns in a collection of images and achieve clinically comparable levels of accuracy in steatosis grading. However, variations in patient populations, acquisition protocols, equipment, and operator expertise across clinical sites can introduce domain shifts that reduce model performance when applied outside the original training setting. In response, unsupervised domain adaptation techniques are being investigated to address these shifts, allowing models to generalize more effectively across diverse clinical environments. In this work, we propose a test-time batch normalization (TTN) technique designed to handle domain shift, especially for changes in label distribution, by adapting selected features of batch normalization (BatchNorm) layers in a trained convolutional neural network model. This approach operates in an unsupervised manner, allowing robust adaptation to new distributions without access to label data. The method was evaluated on two abdominal US datasets collected at different institutions, assessing its capability in mitigating domain shift for hepatic steatosis classification. The proposed method reduced the mean absolute error in steatosis grading by 37% and improved the area under the receiver operating characteristic curves (AUC) for steatosis detection from 0.78 to 0.97, compared to nonadapted models. These findings demonstrate the potential of the proposed method to address domain shift in US-based hepatic steatosis diagnosis, minimizing risks associated with deploying trained models in various clinical settings.
Collapse
|
4
|
Park JH, Hong JY, Han K, Kang W, Shen JJ. Increased Risk of Early-Onset Endometrial Cancer in Women Aged 20-39 Years with Non-Alcoholic Fatty Liver Disease: A Nationwide Cohort Study. Cancers (Basel) 2025; 17:1322. [PMID: 40282498 PMCID: PMC12025614 DOI: 10.3390/cancers17081322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Given the rising incidence of early-onset endometrial cancer diagnosed before the age of 50 years, this study examined whether non-alcoholic fatty liver disease (NAFLD) served as an independent risk factor for early-onset endometrial cancer, irrespective of obesity status. Methods: This nationwide cohort study included 2,311,949 Korean women aged 20-39 years who underwent health screenings from 2009 to 2012. NAFLD severity was classified based on the fatty liver index: none (<30), moderate (30-59), and severe (≥60). Multivariable-adjusted Cox proportional hazards models were applied to estimate adjusted hazard ratios (aHRs) for early-onset endometrial cancer. Results: During a median follow-up of 7.6 years, 1289 women developed early-onset endometrial cancer. The cumulative incidence of early-onset endometrial cancer was significantly higher in women with NAFLD than in those without (log-rank p < 0.001). A dose-dependent association was observed, with increased risk corresponding to greater NAFLD severity (aHR [95% confidence interval (CI)]: moderate NAFLD, 2.38 [1.99-2.85]; severe NAFLD, 5.39 [4.44-6.53]; p for trend < 0.01). Compared with non-obese women without NAFLD, the aHRs for early-onset endometrial cancer were 2.53 [2.11-3.05] in non-obese women with NAFLD, 1.66 [1.10-2.52] in obese women without NAFLD, and 4.30 [3.60-5.13] in obese women with NAFLD (synergy index = 1.50, p < 0.01). Conclusions: NAFLD was independently associated with increased risk of early-onset endometrial cancer in both non-obese and obese women. Furthermore, young women with both NAFLD and obesity exhibited a synergistically elevated risk. Early identification and management of NAFLD may help mitigate the rapidly growing burden of early-onset endometrial cancer.
Collapse
Affiliation(s)
- Joo-Hyun Park
- Department of Family Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Republic of Korea;
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| | - Jung Yong Hong
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 03181, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Wonseok Kang
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology (SAIHST), Sungkyunkwan University, Seoul 03181, Republic of Korea
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jay J. Shen
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas, Las Vegas, NV 89154, USA;
| |
Collapse
|
5
|
Liu Z, Huang J, Dai L, Yuan H, Jiang Y, Suo C, Jin L, Zhang T, Chen X. Steatotic Liver Disease Prevalence in China: A Population-Based Study and Meta-Analysis of 17.4 Million Individuals. Aliment Pharmacol Ther 2025; 61:1110-1122. [PMID: 40013739 DOI: 10.1111/apt.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/20/2024] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Steatotic liver disease (SLD), including metabolic dysfunction-associated SLD (MASLD), has emerged as a leading cause of chronic liver disease in China. AIMS We aimed to provide a comprehensive and updated description of SLD prevalence in China. METHODS We described the prevalence, subgroup distribution, and clinical characteristics of SLD in the Taizhou Study of Liver Diseases (T-SOLID). Additionally, we searched for studies reporting SLD prevalence in five databases. Eligible data were analysed using a generalised linear mixed model. Linear regression was applied to estimate the annual average percentage change (AAPC). RESULTS Of the 28,623 participants in T-SOLID, 30.8% were diagnosed with SLD, among which 83.8% were classified as MASLD. Prevalence of SLD increased from 22.1% in 2018 to 36.7% in 2021. The meta-analysis included 792 publications and 17,404,296 subjects. Nationwide, the pooled SLD prevalence rose from 23.8% (95% CI 21.9%-25.9%) during 2001-2010 to 27.9% (26.0%-29.8%) during 2016-2023 in the general population (AAPC = 2.56, p < 0.0001), equating to approximately 402.0 million cases. An increase in SLD prevalence was observed in subpopulations by region, sex, and age, and in high-risk groups. Northeast China had the highest prevalence (35.0%). Males had a higher prevalence rate than females (35.0% vs. 20.6%). SLD prevalence increased with age, ranging from 8.1% in children and adolescents to 31.8% in the elderly. Meta-regression identified calendar period, age, sex, geographical area, and residence area as significant determinants of SLD prevalence. CONCLUSION The ubiquitously rising prevalence of SLD in Chinese populations underscores the urgent need for targeted public health interventions.
Collapse
Affiliation(s)
- Zhenqiu Liu
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Jiayi Huang
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Luojia Dai
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Huangbo Yuan
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Yanfeng Jiang
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Li Jin
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Xingdong Chen
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Shi W, Wang Y, Chen S, Wei P, Ma D, Zhu J, Zhang Q, Li Z. The association of life's essential 8 scores trajectory patterns with the risk of all cancer types. Sci Rep 2025; 15:9600. [PMID: 40113959 PMCID: PMC11926087 DOI: 10.1038/s41598-025-94009-x] [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: 10/29/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
The "Life's Essential 8" (LE8) is a comprehensive lifestyle assessment tool by the American Heart Association, designed to mitigate cardiovascular disease risk by optimizing lifestyle factors including diet, physical activity, and other health metrics. While individual components of LE8 have been linked to reduced cancer risks, comprehensive studies on LE8 score trajectories over time and their relation to cancer risk are lacking. This study employed the Kailuan cohort, involving 48,330 participants who underwent three health examinations from 2006 to 2010 to determine their LE8 scores. LE8 score trajectories were analyzed using latent mixture modeling, and their association with incident cancer risks was assessed through Cox proportional hazard models, adjusting for confounders such as age, biological sex, and lifestyle behaviors. Three distinct LE8 trajectory patterns were identified: low-stable (21.2%), moderate-stable (49.4%), and elevated-stable (29.4%). Participants with elevated-stable trajectories showed a 21% (HR = 0.79; 95% CI: 0.70-0.90), 27% (HR = 0.73, 95% CI: 0.57-0.92), 51% (HR = 0.49, 95% CI: 0.32-0.77), 31% (HR = 0.69, 95% CI: 0.50-0.97) and 39% (HR = 0.61, 95% CI: 0.39-0.95) reduction in overall, lung, breast, colorectal and liver cancer risk compared to those with low-stable scores. Notably, the protective effect of elevated-stable LE8 scores against breast cancer was pronounced in participants with elevated CRP levels, indicating an interaction between inflammation and LE8 trajectories. Maintaining high and stable LE8 scores significantly associated with reduced cancer risk, underscoring the importance of integrating LE8 into public health and clinical strategies for cancer prevention. Kailuan study, ChiCTR-TNRC-11,001,489. Registered August 24, 2011-Retrospectively registered, http://www.chictr.org.cn/showprojen.aspx?proj=8050 .
Collapse
Affiliation(s)
- Wenzai Shi
- Department of Hepatobiliary Surgery, Peking University International Hospital, Beijing, 102206, China
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, 100044, China
| | - Yiming Wang
- Department of Hepatological Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, Tangshan, 063000, China
| | - Pengcheng Wei
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, 100044, China
| | - Delin Ma
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, 100044, China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, 100044, China
| | - Qingsong Zhang
- Department of General Surgery, Kailuan General Hospital, Tangshan, 063000, China.
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, 100044, China.
- Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
7
|
Shi W, Suo X, Wang Y, Yao N, Xing S, Wei P, Ma D, Zhu J, Zhang Q, Li Z. The associations between irregular breakfast and late-night snacking with metabolic dysfunction-associated steatotic liver disease. Clin Nutr 2025; 46:52-59. [PMID: 39864380 DOI: 10.1016/j.clnu.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND The newly coined term Metabolic dysfunction-associated steatotic liver disease (MASLD) emphasizes the critical role of metabolic risk factors in the pathogenesis of fatty liver disease. The consumption of irregular breakfasts or late-night snacks has been identified as a factor closely associated with disruptions in the body's energy homeostasis and metabolic balance. However, the relationship between these behaviors and MASLD has not been previously examined. METHODS Participants in this study were recruited from the Kailuan cohort, a prospective cohort. All participants completed questionnaires regarding breakfast and late-night snack frequencies. The primary outcomes were MASLD and MASLD with increased alcohol intake (MetALD). Cox regression analysis was utilized to calculate the hazard ratios (HR) and 95 % confidence intervals (CI) for MASLD and MetALD in different groups. Mediation analysis was used to assess the mediating effect of BMI on this relationship. RESULTS A total of 32,030 participants were included in the study. Both irregular breakfast consumption and late-night snacking were associated with an increased risk of MASLD. Compared to participants who consumed regular breakfast and no late-night snacks, the risk of MASLD was elevated in participants who consumed regular breakfast but late-night snacks (HR = 1.12, 95 % CI: 1.05-1.20), irregular breakfast but no late-night snacking (HR = 1.16, 95 % CI: 1.05-1.27), irregular breakfast and late-night snacking (HR = 1.26, 95 % CI: 1.13-1.42), never ate breakfast and no late-night snacking (HR = 1.33, 95 % CI: 1.23-1.44), and never ate breakfast and late-night snacking (HR = 1.52, 95 % CI: 1.30-1.77). CONCLUSION Irregular or skipped breakfast and late-night snacking were associated with an increased risk of MASLD and MetALD.
Collapse
Affiliation(s)
- Wenzai Shi
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China; Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, China; Department of Hepatobiliary Surgery, Peking University International Hospital, Beijing, China
| | - Xiaopeng Suo
- Department of General Surgery, Peking University People's Hospital, Beijing, China
| | - Yiming Wang
- Department of General Surgery, Kailuan General Hospital, Tangshan, China
| | - Nan Yao
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Siyu Xing
- Department of Biology, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Pengcheng Wei
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China; Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, China
| | - Delin Ma
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China; Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China; Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, China
| | - Qingsong Zhang
- Department of General Surgery, Kailuan General Hospital, Tangshan, China.
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China; Beijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
8
|
Åström H, Takami Lageborn C, Hagström H. Psychosocial risks in metabolic dysfunction-associated steatotic liver disease. Expert Rev Gastroenterol Hepatol 2025:1-18. [PMID: 39953908 DOI: 10.1080/17474124.2025.2468297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/03/2025] [Accepted: 02/13/2025] [Indexed: 02/17/2025]
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) is increasingly becoming more prevalent in the general population. MASLD is more common in persons with low socioeconomic status (SES), yet little is known about the psychosocial challenges associated with this disease, and clinical recommendations on how to approach psychosocial challenges are lacking. AREAS COVERED A PubMed search using the search terms MASLD, psychosocial risks, stigmatization, psychiatric comorbidities (i.e. depression, bipolar disorder, psychosis, attention deficit hyperactivity disorder, and substance abuse), SES, quality of life (QoL), over the past 20 years (2004-2024) was performed. EXPERT OPINION Persons with MASLD often experience psychosocial adversities that may be expressed as lower SES, high prevalence of depression, and reduced QoL. Knowledge gaps remain regarding the association between severe mental disorders (e.g. psychosis and bipolar disorders). Timely detection and treatment of MASLD in persons with psychosocial risks may require attention and cross-field collaboration. Studies on QoL in persons with MASLD differ in methodology which makes formal comparisons difficult. Psychosocial adversity may be a barrier to lifestyle modifications, which remain the cornerstone of MASLD management. Guidelines on how to address psychosocial adversities in a clinical setting are warranted to improve outcomes and decrease further multimorbidity.
Collapse
Affiliation(s)
- Hanne Åström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Hannes Hagström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Liu K, Wang W, Liu Y, Li J, Ma C, Tian Y, Dong Z, Zhu L, Wei W, Ren M, Wu S, Liu S. Correlation of cumulative fasting blood glucose exposure with gastrointestinal cancers: A prospective cohort study. Medicine (Baltimore) 2025; 104:e41529. [PMID: 39960953 PMCID: PMC11835132 DOI: 10.1097/md.0000000000041529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
At present, there is a lack of research on the correlation between cumFPG and digestive malignancies, and previous cohort studies have not considered the competitive risk between death and digestive malignancies, which may overestimate the impact of related risk factors. To explore the correlation between cumFPG and malignant tumors of the digestive system. In this study, 53,747 participants who had undergone 3 consecutive physical examinations since 2006 were collected. Finally, a total of 53,747 participants were included in this study. According to the grouping method of previous studies, cumFPG was divided into 4 groups according to the quartile. Cox regression model and competitive risk model were used to assess the risk of new digestive system malignancy. In sensitivity analyses, participants with cancer within 5 years of follow-up were excluded to eliminate the possibility of reverse causation. Subjects taking hypoglycemic drugs were excluded to eliminate the effect of the drug on blood glucose. Restricted cubic splineregresion (RCS) was then used to calculate the relationship between cumFPG and GI cancers. The mean age of participants was 49.02 ± 11.78 years. During a mean follow-up of 10.58 years, 817 new Gastrointestinal cases were identified, and the Cox proportional hazards model suggested that the risk of incidence in the Q2 to Q4 group increased sequentially compared with the lowest Q1 group, even after excluding the diagnosis of digestive malignancy within 5 years, the participants taking hypoglycemic drugs, and the death competition risk model analysis. In site-specific analysis, we observed that this risk was more pronounced in colorectal cancer, liver cancer, and pancreatic cancer, while gastric cancer, small bowel cancer, and bile duct cancer all had a similar trend to the main model but were not statistically significant, while esophageal cancer was U-shaped but not statistically significant. RCS results showed that cumFPG was associated with a similar risk of digestive system tumors, showing an inverted "√" type relationship. High levels of cumFPG are an independent factor in malignancy of the digestive system. cumFPG can provide a new idea for the prevention of Gastrointestinal cancers.
Collapse
Affiliation(s)
- Kuan Liu
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Wanchao Wang
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Ye Liu
- Affiliated Hospital of North China University of Science and Technology, Breast Disease Treatment Center, Tangshan, Hebei, China
| | - Jiaxing Li
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Chao Ma
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Yuan Tian
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Zhigang Dong
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Lichao Zhu
- Affiliated Hospital of North China University of Science and Technology, GastrointestinalOncology Treatment Center, Tangshan, Hebei, China
| | - Wenqiang Wei
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Minqiang Ren
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| | - Shouling Wu
- Kailuan Employee Health Examination Center, Tangshan, Hebei, China
| | - Siqing Liu
- Affiliated Hospital of North China University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, Tangshan, Hebei, China
| |
Collapse
|
10
|
Danpanichkul P, Suparan K, Prasitsumrit V, Ahmed A, Wijarnpreecha K, Kim D. Long-term outcomes and risk modifiers of metabolic dysfunction-associated steatotic liver disease between lean and non-lean populations. Clin Mol Hepatol 2025; 31:74-89. [PMID: 39439408 PMCID: PMC11791619 DOI: 10.3350/cmh.2024.0631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024] Open
Abstract
One-third of adults across the globe exhibit metabolic dysfunction-associated steatotic liver disease (MASLD)-formerly known as nonalcoholic fatty liver disease (NAFLD). To date, MASLD is the fastest-growing etiology of chronic liver disease and hepatocellular carcinoma (HCC). Besides the population with obesity, MASLD can also be found in lean populations, accounting for 13% of the global population, especially Asians. Notably, individuals with lean MASLD face equal or higher overall mortality rates compared to their non-lean counterparts. Risk modifiers encompass advanced age, hepatic fibrosis, and type 2 diabetes mellitus (T2DM). Moreover, the population with lean MASLD is associated with an increased risk of HCC, while their non-lean counterparts are more prone to cardiovascular outcomes and T2DM. Existing evidence indicates a similar risk of liver-related events and extrahepatic cancer between the two groups. However, MASLD-related genetic variants, such as PNPLA3 and TM6SF2, did not significantly affect mortality between the two populations. Still, underreporting alcohol consumption and regional representation limits the study's comprehensiveness. Longitudinal studies and mechanistic explorations are needed to understand differences in lean versus non-lean MASLD populations. This review highlights the need for awareness and tailored interventions in managing MASLD, considering lean individuals' unique risks.
Collapse
Affiliation(s)
- Pojsakorn Danpanichkul
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kanokphong Suparan
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Aijaz Ahmed
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Karn Wijarnpreecha
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Banner University Medical Center, Phoenix, AZ, USA
- BIO5 Institute, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Donghee Kim
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Perry AS, Hadad N, Chatterjee E, Jimenez-Ramos M, Farber-Eger E, Roshani R, Stolze LK, Betti MJ, Zhao S, Huang S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Vaitinadin NS, Freedman JE, Tanriverdi K, Alsop E, Van Keuren-Jensen K, Sauld JFK, Mahajan G, Khan SS, Colangelo L, Nayor M, Fisher-Hoch S, McCormick JB, North KE, Below JE, Wells QS, Abel ED, Kalhan R, Scott C, Guilliams M, Gamazon ER, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. Cell Rep Med 2024; 5:101871. [PMID: 39657669 PMCID: PMC11722105 DOI: 10.1016/j.xcrm.2024.101871] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Hepatic steatosis is a central phenotype in multi-system metabolic dysfunction and is increasing in parallel with the obesity pandemic. We use a translational approach integrating clinical phenotyping and outcomes, circulating proteomics, and tissue transcriptomics to identify dynamic, functional biomarkers of hepatic steatosis. Using multi-modality imaging and broad proteomic profiling, we identify proteins implicated in the progression of hepatic steatosis that are largely encoded by genes enriched at the transcriptional level in the human liver. These transcripts are differentially expressed across areas of steatosis in spatial transcriptomics, and several are dynamic during stages of steatosis. Circulating multi-protein signatures of steatosis strongly associate with fatty liver disease and multi-system metabolic outcomes. Using a humanized "liver-on-a-chip" model, we induce hepatic steatosis, confirming cell-specific expression of prioritized targets. These results underscore the utility of this approach to identify a prognostic, functional, dynamic "liquid biopsy" of human liver, relevant to biomarker discovery and mechanistic research applications.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Niran Hadad
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Maria Jimenez-Ramos
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Michael J Betti
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shilin Zhao
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shi Huang
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Timothy J Kendall
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK; Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Tinne Thone
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Samuel Bailin
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L Gabriel
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J Jeffrey Carr
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Jane E Freedman
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Eric Alsop
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | | | - Sadiya S Khan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Laura Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Susan Fisher-Hoch
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Joseph B McCormick
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Kari E North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quinn S Wells
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ravi Kalhan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Charlotte Scott
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Eric R Gamazon
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Ravi Shah
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
12
|
Lan T, Tacke F. Diagnostics and omics technologies for the detection and prediction of metabolic dysfunction-associated steatotic liver disease-related malignancies. Metabolism 2024; 161:156015. [PMID: 39216799 DOI: 10.1016/j.metabol.2024.156015] [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: 06/01/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise, making it the leading etiology of chronic liver diseases and a prime cause of liver-related mortality. MASLD can progress into steatohepatitis (termed MASH), fibrosis, cirrhosis, and ultimately cancer. MASLD is associated with increased risks of hepatocellular carcinoma (HCC) and also extrahepatic malignancies, which can develop in both cirrhotic and non-cirrhotic patients, emphasizing the importance of identifying patients with MASLD at risk of developing MASLD-associated malignancies. However, the optimal screening, diagnostic, and risk stratification strategies for patients with MASLD at risk of cancer are still under debate. Individuals with MASH-associated cirrhosis are recommended to undergo surveillance for HCC (e.g. by ultrasound and biomarkers) every six months. No specific screening approaches for MASLD-related malignancies in non-cirrhotic cases are established to date. The rapidly developing omics technologies, including genetics, metabolomics, and proteomics, show great potential for discovering non-invasive markers to fulfill this unmet need. This review provides an overview on the incidence and mortality of MASLD-associated malignancies, current strategies for HCC screening, surveillance and diagnosis in patients with MASLD, and the evolving role of omics technologies in the discovery of non-invasive markers for the prediction and risk stratification of MASLD-associated HCC.
Collapse
Affiliation(s)
- Tian Lan
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany; Laboratory of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| |
Collapse
|
13
|
Mantovani A, Lonardo A, Stefan N, Targher G. Metabolic dysfunction-associated steatotic liver disease and extrahepatic gastrointestinal cancers. Metabolism 2024; 160:156014. [PMID: 39182602 DOI: 10.1016/j.metabol.2024.156014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/09/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) poses a significant and ever-increasing health and economic burden worldwide. Substantial epidemiological evidence shows that MASLD is a multisystem disease that is associated not only with liver-related complications but is also associated with an increased risk of developing cardiometabolic comorbidities and extrahepatic cancers (principally gastrointestinal [GI] cancers). GI cancers account for a quarter of the global cancer incidence and a third of cancer-related deaths. In this narrative review, we provide an overview of the literature on (a) the epidemiological data on the risk of non-liver GI cancers in MASLD, (b) the putative mechanisms by which MASLD (and factors linked with MASLD) may increase this risk, and (c) the possible pharmacotherapies beneficially affecting both MASLD and extrahepatic GI cancer risk. There are multiple potential pathophysiological mechanisms by which MASLD may increase extrahepatic GI cancer risk. Although further studies are needed, the current evidence supports a possible extrahepatic carcinogenic role for MASLD, regardless of obesity and diabetes status, thus highlighting the potential role of tailoring cancer screening for individuals with MASLD. Although there are conflicting data in the literature, aspirin, statins and metformin appear to exert some chemo-preventive effects against GI cancer.
Collapse
Affiliation(s)
- Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Amedeo Lonardo
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Norbert Stefan
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tübingen, Germany
| | - Giovanni Targher
- Department of Medicine, University of Verona, Italy; Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy.
| |
Collapse
|
14
|
Ma T, Hao XM, Zhang X, Liu XY, Wang YM, Zhang QS, Zhang J. In utero and childhood exposure to the great Chinese famine and risk of aging in adulthood. Sci Rep 2024; 14:25089. [PMID: 39443668 PMCID: PMC11499915 DOI: 10.1038/s41598-024-77283-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024] Open
Abstract
Background Early-life exposure to famine may influence the occurrence of chronic diseases and aging in midlife among those exposed. This study aims to explore the relationship between exposure to the Chinese Great Famine and aging in middle-aged individuals. Methods Participants born in 1963-1965 (unexposed), 1959-1961 (in utero exposure), and 1955-1957 (childhood exposure) from the Kailuan Study were included. Their biological age at 2010, 2014, and 2018 was investigated, and age acceleration (biological age minus actual age) was calculated to assess aging. Logistic regression analysis was employed to describe the relationship between famine exposure and the aging risk. Subgroup and sensitivity analysis were conducted to explore differences and stability in this relationship among different groups. Results A total of 17,543 participants were included in this study. Among them, 12,762 (72.7%) were male, and 4,781 (27.3%) were female, with 2,543 participants experiencing aging events. Compared to unexposed participants, those exposed during childhood and in utero exhibited a 1.69-fold (OR = 1.69, 95%CI: 1.53-1.87) and 1.22-fold (OR = 1.22, 95%CI: 1.08-1.37) increased risk of aging. Subgroup analysis revealed an interaction with income (P for interaction = 0.008), and additional interaction analysis suggested that increasing income could partially mitigate the detrimental effects of early-life famine exposure. Furthermore, experiencing famine in severely affected regions exacerbated the risk of aging (OR = 1.41, 95%CI: 1.21-1.63). Conclusion Exposure to famine in utero or during childhood may elevate the risk of midlife aging among exposed individuals, and these relationships are influenced by the severity of famine exposure. Increasing income may also help mitigate these effects.Trial registration: Kailuan study, ChiCTRTNRC11001489. Registered July 19, 2015 Retrospectively registered, https//www.chictr.org.cn/showprojEN.html?proj=8050&u_atoken=af46a0dee8d73f320bb5459ab7bbcfa9&u_asig=1a0c381017255295896468605e00cf .
Collapse
Affiliation(s)
- Tao Ma
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiao-Meng Hao
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaowei Zhang
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin-Yu Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yi-Ming Wang
- Department of Hepatological Surgery, Kailuan General Hospital, Tangshan, China
| | - Qing-Song Zhang
- Department of General Surgery, Kailuan General Hospital, Tangshan, 063000, China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
15
|
He L, Zheng W, Liao Y, Kong W, Zeng T. Individuals with cardiometabolic risk factors are at higher risk for early-onset NAFLD. J Hepatol 2024; 81:e99-e101. [PMID: 38703832 DOI: 10.1016/j.jhep.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024]
Affiliation(s)
- Linfeng He
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China
| | - Wenbin Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China
| | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China.
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China.
| |
Collapse
|
16
|
Kim J, Seki E. Unveiling the cancer risk nexus of the steatotic liver. Trends Endocrinol Metab 2024; 35:708-719. [PMID: 38531699 PMCID: PMC11321945 DOI: 10.1016/j.tem.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
Steatotic liver, characterized by the accumulation of fat in the liver, poses significant health risks including metabolic dysfunction-associated steatotic liver disease (MASLD) and an elevated risk of primary liver cancer. Emerging evidence indicates a robust association between steatotic liver and increased susceptibility to extrahepatic primary cancers and their metastases. The deposition of fat induces dynamic changes in hepatic microenvironments, thereby fostering inflammation and immune responses that enhance liver metastasis from extrahepatic primary cancers. This review explores the impact of steatotic liver on hepatic carcinogenesis and metastasis from extrahepatic cancers, with a specific focus on hepatocyte-derived factors and the immune microenvironment. By emphasizing novel conclusions, this article underscores the timely relevance of understanding these intricate connections.
Collapse
Affiliation(s)
- Jieun Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
17
|
Santoro S, Khalil M, Abdallah H, Farella I, Noto A, Dipalo GM, Villani P, Bonfrate L, Di Ciaula A, Portincasa P. Early and accurate diagnosis of steatotic liver by artificial intelligence (AI)-supported ultrasonography. Eur J Intern Med 2024; 125:57-66. [PMID: 38490931 DOI: 10.1016/j.ejim.2024.03.004] [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: 12/22/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
OBJECTIVES Steatotic liver disease is the most frequent chronic liver disease worldwide. Ultrasonography (US) is commonly employed for the assessment and diagnosis. Few information is available on the possible use of artificial intelligence (AI) to ameliorate the diagnostic accuracy of ultrasonography. MATERIALS AND METHODS An AI-based algorithm was developed using a dataset of US images. We prospectively enrolled 134 patients for algorithm validation. Patients underwent abdominal US and Proton Density Fat Fraction MRI scans (MRI-PDFF), assumed as reference technique. The hepatorenal index was manually calculated (HRIM) by 4 operators. An automatic hepatorenal index (HRIA) was obtained by the algorithm. The accuracy of HRIA to discriminate steatosis grades was evaluated by ROC analysis using MRI-PDFF cut-offs. RESULTS Overweight was 40 % of subjects (BMI 26.4 kg/cm2). The median HRIA was 1.11 (IQR 0.32) and the average of 4 manually calculated HRIM was 1.08 (IQR 0.26), with a 15 % inter-operator variability. Both HRIA (R = 0.79, P < 0.0001) and HRIM (R = 0.69, P < 0.0001) significantly correlated with liver fat percentage (MRI-PDFF). According to MRI-PDFF, 32 % of enrolled subjects had steatosis. Discrimination capacity by AUC between patient with steatosis and patient without steatosis was better for HRIA than HRIM (AUC: 0.87 vs. 0.82, respectively). ROC analysis showed an AUC = 0.98 for HRIA with 1.64 cut-off in distinguishing between mild and moderate/severe groups. CONCLUSIONS The use of AI improves accuracy and speed of ultrasonography in the diagnosis of liver steatosis. Further studies should evaluate the routine use of this technique in the management of liver steatosis at high cardio-metabolic risk.
Collapse
Affiliation(s)
- Sergio Santoro
- PhD Program in Public Health, Clinical Medicine and Oncology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy; Eurisko Technology srl, Modugno, BA, Italy
| | - Mohamad Khalil
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Hala Abdallah
- PhD Program in Public Health, Clinical Medicine and Oncology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy; Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Ilaria Farella
- PhD Program in Public Health, Clinical Medicine and Oncology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy; Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Antonino Noto
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Bari, Italy
| | | | | | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Agostino Di Ciaula
- PhD Program in Public Health, Clinical Medicine and Oncology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy; Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Piero Portincasa
- PhD Program in Public Health, Clinical Medicine and Oncology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy.
| |
Collapse
|
18
|
Deng L, Liu T, Liu CA, Zhang Q, Song MM, Lin SQ, Wang YM, Zhang QS, Shi HP. The association of metabolic syndrome score trajectory patterns with risk of all cancer types. Cancer 2024; 130:2150-2159. [PMID: 38462898 DOI: 10.1002/cncr.35235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/05/2024] [Accepted: 01/22/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Metabolic syndrome (MetS) elevates cancer risk. However, a single MetS assessment does not fully reveal the long-term association with cancer. Inflammation, alongside MetS, could synergistically expedite both the onset and advancement of cancer. This study aims to investigate MetS score trajectories and cancer risk in a large, prospective cohort study. METHODS The authors prospectively examined the relationship between MetS score trajectory patterns and new-onset cancer in 44,115 participants. Latent mixture modeling was used to identify the MetS score trajectories. Cox proportional hazards regression models were used to evaluate the association between MetS score trajectory patterns and the risk of overall and site-specific cancers. RESULTS Four MetS score trajectory patterns were identified: low-stable (n = 4657), moderate-low (n = 18,018), moderate-high (n = 18,288), and elevated-increasing (n = 3152). Compared to participants with a low-stable trajectory pattern, the elevated-increasing trajectory pattern was associated with an elevated risk of overall (hazard ratio [HR], 1.27; 95% confidence interval [CI], 1.04-1.55), breast (HR, 2.11; 95% CI, 1.04-4.34), endometrial (HR, 3.33; 95% CI, 1.16-6.77), kidney (HR, 4.52; 95% CI, 1.17-10.48), colorectal (HR, 2.54; 95% CI, 1.27-5.09), and liver (HR, 1.61; 95% CI, 1.09-4.57) cancers. Among participants with chronic inflammation (C-reactive protein levels ≥3 mg/L), the elevated-increasing trajectory pattern was significantly associated with subsequent breast, endometrial, colorectal, and liver cancers. CONCLUSIONS Trajectories of MetS scores are associated with the occurrence of cancers, especially breast, endometrial, kidney, colorectal, and liver cancers, emphasizing the importance of long-term monitoring and evaluation of MetS. PLAIN LANGUAGE SUMMARY The association between long-term elevated metabolic syndrome (MetS) scores and a heightened risk of various cancers is a pivotal finding of our study. Our research further indicates that individuals with MetS, particularly when coupled with chronic inflammation, are at an increased risk of cancer. We propose that sustained monitoring and management of MetS could be beneficial in reducing cancer risk.
Collapse
Affiliation(s)
- Li Deng
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tong Liu
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chen-An Liu
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Qi Zhang
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Meng-Meng Song
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Shi-Qi Lin
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yi-Ming Wang
- Department of Hepatological Surgery, Kailuan General Hospital, Tangshan, China
| | - Qing-Song Zhang
- Department of General Surgery, Kailuan General Hospital, Tangshan, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Di Ciaula A, Portincasa P. Bridging current knowledge gap: better primary colorectal cancer prevention in people living with metabolic dysfunction-associated steatotic liver. Intern Emerg Med 2024; 19:887-889. [PMID: 38691255 DOI: 10.1007/s11739-024-03617-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePrev-J), University of Bari Medical School, P.zza Giulio Cesare 11, 70124, Bari, Italy.
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Precision and Regenerative Medicine and Ionian Area (DiMePrev-J), University of Bari Medical School, P.zza Giulio Cesare 11, 70124, Bari, Italy
| |
Collapse
|
20
|
Yeh JG, Newsome PN. Association of glucagon-like peptide-1 receptor agonists with serious liver events among patients with type 2 diabetes: A Scandinavian cohort study. Hepatology 2024; 79:1255-1257. [PMID: 38345826 DOI: 10.1097/hep.0000000000000768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 04/05/2024]
Affiliation(s)
- Julian G Yeh
- Department of Liver Medicine, Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK
- Department of Immunology and Immunotherapy, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Philip N Newsome
- Department of Liver Medicine, Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK
- Department of Immunology and Immunotherapy, Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
21
|
Kokkorakis M, Muzurović E, Volčanšek Š, Chakhtoura M, Hill MA, Mikhailidis DP, Mantzoros CS. Steatotic Liver Disease: Pathophysiology and Emerging Pharmacotherapies. Pharmacol Rev 2024; 76:454-499. [PMID: 38697855 DOI: 10.1124/pharmrev.123.001087] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 05/05/2024] Open
Abstract
Steatotic liver disease (SLD) displays a dynamic and complex disease phenotype. Consequently, the metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) therapeutic pipeline is expanding rapidly and in multiple directions. In parallel, noninvasive tools for diagnosing and monitoring responses to therapeutic interventions are being studied, and clinically feasible findings are being explored as primary outcomes in interventional trials. The realization that distinct subgroups exist under the umbrella of SLD should guide more precise and personalized treatment recommendations and facilitate advancements in pharmacotherapeutics. This review summarizes recent updates of pathophysiology-based nomenclature and outlines both effective pharmacotherapeutics and those in the pipeline for MASLD/MASH, detailing their mode of action and the current status of phase 2 and 3 clinical trials. Of the extensive arsenal of pharmacotherapeutics in the MASLD/MASH pipeline, several have been rejected, whereas other, mainly monotherapy options, have shown only marginal benefits and are now being tested as part of combination therapies, yet others are still in development as monotherapies. Although the Food and Drug Administration (FDA) has recently approved resmetirom, additional therapeutic approaches in development will ideally target MASH and fibrosis while improving cardiometabolic risk factors. Due to the urgent need for the development of novel therapeutic strategies and the potential availability of safety and tolerability data, repurposing existing and approved drugs is an appealing option. Finally, it is essential to highlight that SLD and, by extension, MASLD should be recognized and approached as a systemic disease affecting multiple organs, with the vigorous implementation of interdisciplinary and coordinated action plans. SIGNIFICANCE STATEMENT: Steatotic liver disease (SLD), including metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis, is the most prevalent chronic liver condition, affecting more than one-fourth of the global population. This review aims to provide the most recent information regarding SLD pathophysiology, diagnosis, and management according to the latest advancements in the guidelines and clinical trials. Collectively, it is hoped that the information provided furthers the understanding of the current state of SLD with direct clinical implications and stimulates research initiatives.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Emir Muzurović
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Špela Volčanšek
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Marlene Chakhtoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Michael A Hill
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Dimitri P Mikhailidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| |
Collapse
|
22
|
Li Y, Yang P, Ye J, Xu Q, Wu J, Wang Y. Updated mechanisms of MASLD pathogenesis. Lipids Health Dis 2024; 23:117. [PMID: 38649999 PMCID: PMC11034170 DOI: 10.1186/s12944-024-02108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has garnered considerable attention globally. Changing lifestyles, over-nutrition, and physical inactivity have promoted its development. MASLD is typically accompanied by obesity and is strongly linked to metabolic syndromes. Given that MASLD prevalence is on the rise, there is an urgent need to elucidate its pathogenesis. Hepatic lipid accumulation generally triggers lipotoxicity and induces MASLD or progress to metabolic dysfunction-associated steatohepatitis (MASH) by mediating endoplasmic reticulum stress, oxidative stress, organelle dysfunction, and ferroptosis. Recently, significant attention has been directed towards exploring the role of gut microbial dysbiosis in the development of MASLD, offering a novel therapeutic target for MASLD. Considering that there are no recognized pharmacological therapies due to the diversity of mechanisms involved in MASLD and the difficulty associated with undertaking clinical trials, potential targets in MASLD remain elusive. Thus, this article aimed to summarize and evaluate the prominent roles of lipotoxicity, ferroptosis, and gut microbes in the development of MASLD and the mechanisms underlying their effects. Furthermore, existing advances and challenges in the treatment of MASLD were outlined.
Collapse
Affiliation(s)
- Yuxuan Li
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Peipei Yang
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jialu Ye
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Qiyuan Xu
- Wenzhou Medical University, Wenzhou, China
| | - Jiaqi Wu
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
| | - Yidong Wang
- Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
23
|
He L, Qiu K, Zheng W, Kong W, Zeng T. Uric acid may serve as the sixth cardiometabolic criterion for defining MASLD. J Hepatol 2024; 80:e152-e153. [PMID: 38110009 DOI: 10.1016/j.jhep.2023.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/03/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023]
Affiliation(s)
- Linfeng He
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China
| | - Kangli Qiu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China
| | - Wenbin Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China.
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, China.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW This review aims to discuss the most recent evidence exploring the role of lipid droplets in steatotic liver disease (SLD). We highlight the breadth of mechanisms by which lipid droplets may contribute to the progression of SLD with a particular focus on the role of lipid droplets as inducers of mechanical stress within hepatocytes and genetic mutations in lipid droplet associated proteins. Finally, this review provides an update on clinical trials exploring the therapeutic potential and strategies targeting lipid droplets. RECENT FINDINGS The size, composition and location of hepatic lipid droplets strongly influence the pathological role of these organelles in SLD. Emerging studies are beginning to elucidate the importance of lipid droplet induced hepatocyte mechanical stress. Novel strategies targeting lipid droplets, including the effects of lipid droplet associated protein mutations, show promising therapeutic potential. SUMMARY Much more than a histological feature, lipid droplets are complex heterogenous organelles crucial to cellular metabolism with important causative roles in the development and progression of SLD. Lipid droplet induced mechanical stress may exacerbate hepatic inflammation and fibrogenesis and potentially contribute to the development of a pro-carcinogenic hepatic environment. The integration of advancements in genetics and molecular biology in upcoming treatments aspires to transcend symptomatic alleviation and address the fundamental causes and pathological development of SLD.
Collapse
Affiliation(s)
- Josh Bilson
- School of Human Development and Health, Faculty of Medicine, University of Southampton
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
| | - Eleonora Scorletti
- School of Human Development and Health, Faculty of Medicine, University of Southampton
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Liu K, Tang S, Liu C, Ma J, Cao X, Yang X, Zhu Y, Chen K, Liu Y, Zhang C, Liu Y. Systemic immune-inflammatory biomarkers (SII, NLR, PLR and LMR) linked to non-alcoholic fatty liver disease risk. Front Immunol 2024; 15:1337241. [PMID: 38481995 PMCID: PMC10933001 DOI: 10.3389/fimmu.2024.1337241] [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: 11/12/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024] Open
Abstract
Background Systemic immune-inflammatory biomarkers including systemic immune inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) have been demonstrated to be associated with the risk and severity of various liver diseases. However, studies on their role and clinical significance in metabolic diseases, especially in nonalcoholic fatty liver disease (NAFLD), are limited and results are inconsistent. Methods 10821 adults aged 20 years or older were enrolled in this cross-sectional study, sourced from six cycles of the National Health and Nutrition Examination Survey (NHANES). Survey-weighted logistic regression was employed to investigate the correlation between systemic immune-inflammatory biomarkers (SII, NLR, PLR, and LMR) and NAFLD risk. Restricted cubic spline regression models and segmented regression models were used to describe nonlinear relationships and threshold effects. Subgroup and sensitivity analyses were also conducted. Results After adjusting for all confounding variables, there was a significant positive association observed between ln-transformed SII (OR= 1.46, 95% CI: 1.27-1.69, P <0.001), NLR (OR= 1.25, 95% CI: 1.05-1.49, P =0.015), LMR (OR= 1.39, 95% CI: 1.14-1.69, P = 0.002) with NAFLD. A nonlinear dose-response relationship with an inverted "U"-shaped threshold of 4.64 was observed between ln(PLR) and NAFLD risk. When ln(PLR) was below 4.64, each unit increase in ln(PLR) was associated with a 0.55-fold increase in the risk of NAFLD (OR= 1.55, 95% CI: 1.05-2.31, P <0.05). Conversely, when ln(PLR) exceeded 4.64, each unit increase in ln(PLR) was associated with a 0.40-fold decrease in the risk of NAFLD (OR= 0.60, 95% CI. 0.44-0.81, P <0.05). Conclusion ln-transformed SII, NLR, and LMR were linearly associated with NAFLD risk. ln(PLR) showed an inverted "U"-shaped nonlinear dose-response relationship with the risk of NAFLD.
Collapse
Affiliation(s)
- Ke Liu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shiyun Tang
- The National Clinical Trial Center of Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Chenhao Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jianli Ma
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiyu Cao
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiuli Yang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi Zhu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ke Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- People's Hospital of Xinjin District, Chengdu, Sichuan, China
| | - Ya Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Liu C, Liu T, Zhang Q, Song M, Zhang Q, Shi J, Deng L, Chen Y, Zheng X, Lin S, Wang Z, Xie H, Chen S, Wu S, Shi H. Temporal relationship between inflammation and metabolic disorders and their impact on cancer risk. J Glob Health 2024; 14:04041. [PMID: 38386717 PMCID: PMC10869135 DOI: 10.7189/jogh.14.04041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Background Inflammation and metabolic disorders are closely associated with cancer. Whether inflammation leads to metabolic disorders or vice versa during cancer initiation remains unclear. In this study, we explored this temporal relationship and the co-exposure effect on cancer risk. Methods This prospective study had two phases. Initially, we examined the temporal relationship between inflammation (high-sensitivity C-reactive protein (CRP)) and metabolic disorders (metabolic syndrome severity Z-score (MetS-Z)) using a 3.98-year survey and cross-lagged analysis. Subsequently, we assessed the connection of co-exposure to inflammation and metabolic disorders, and the risks of overall cancer, as well as specific obesity-related, non-obesity-related, digestive system, lung, and other cancers using an 11.04-year survey and Cox proportional hazard models. Results The cross-lagged analysis revealed that the path coefficient from baseline CRP to follow-up MetS-Z (β2 = 0.032; 95% confidence interval (CI) = 0.026, 0.046) was more significant than the path coefficient from baseline MetS-Z to follow-up CRP (β1 = 0.009; 95% CI = -0.001, 0.019). During the follow-up, 2304 cases of cancer occurred. Compared with the risk of cancer of patients with low average cumulative CRP and MetS-Z, patients with high value had a significantly increased risk (hazard ratio = 1.54, 95% CI = 1.30, 1.83). The mediation analysis showed that MetS-Z mediated the association between CRP levels and overall cancer (12.67%), digestive system cancer (10.16%), and obesity-related cancer risk (13.87%). Conclusions Inflammation had a greater impact on metabolic disorders than vice versa. Co-exposure to inflammation and metabolic disorders significantly increased the risk of cancer, particularly digestive system and obesity-related cancers. Registration Chinese Clinical Trial Registry: ChiCTR-TNRC-11001489.
Collapse
Affiliation(s)
- Chenan Liu
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Tong Liu
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Qingsong Zhang
- Department of General Surgery, Kailuan General Hospital, Tangshan, China
| | - Mengmeng Song
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - Qi Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, USA
| | - Jinyu Shi
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Li Deng
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Yue Chen
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Xin Zheng
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Shiqi Lin
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Ziwen Wang
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Hailun Xie
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery, Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
- Laboratory for Clinical Medicine Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Di Ciaula A, Bonfrate L, Portincasa P. The fatty liver as testimonial of systemic diseases. Further evidence from rheumatoid arthritis and confirmation for a leading role of internal medicine. Eur J Intern Med 2023; 118:41-42. [PMID: 37872035 DOI: 10.1016/j.ejim.2023.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica "A. Murri", Department of Preventive and Regenerative Medicine and Ionian Area (DiMePrev-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11 Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Preventive and Regenerative Medicine and Ionian Area (DiMePrev-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11 Bari, Italy
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Preventive and Regenerative Medicine and Ionian Area (DiMePrev-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11 Bari, Italy.
| |
Collapse
|