1
|
Yan JH, Hao PL, Zeng SM, Liang CX, Chen Y, Chen KW, Qu WQ, Zeng X, Zhong Z, Zhang XZ. Injectable multifunctional hydrogels for adipose tissue remodeling to treat obesity and alleviate metabolic syndrome. J Control Release 2025; 381:113585. [PMID: 40032012 DOI: 10.1016/j.jconrel.2025.113585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Dysfunction of adipose tissue in obese individuals is associated with metabolic imbalances. Current treatments focus on reducing calorie intake and increasing exercise but have unsatisfactory efficacy and significant side effects. Here, a subcutaneously in situ formed therapeutic hydrogel (RL lip@gel) that can simultaneously modulate the inflammatory environment of adipose tissue and induce adipocyte browning is developed to combat obesity. The alginate solution encapsulates the anti-inflammatory natural flavonoid luteolin and rosiglitazone-loaded liposomes (RL lip), which can be formed as an in situ hydrogel through crosslinking with divalent metal ions in adipose tissue following subcutaneous injection. RL lip@gel not only reprograms M1-like adipose tissue macrophages into an M2-like phenotype but diminishes the expression of proinflammatory cytokine, thereby decreasing systemic glucose resistance and lipid storage. Meanwhile, rosiglitazone embedded within the adipose tissue promotes the browning of white adipocytes, and boosts energy expenditure, with further enhancement from luteolin, as demonstrated by decreased white adipose tissue masses. This localized intervention not only ensures the efficacy of combating obesity systemically but enhances overall metabolism while minimizing side effects.
Collapse
Affiliation(s)
- Jian-Hua Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Pei-Ling Hao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Si-Min Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Chun-Xiao Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China
| | - Ke-Wei Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Wen-Qiang Qu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| | - Zhenlin Zhong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, PR China; Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
2
|
Eslam M, Fan JG, Yu ML, Wong VWS, Cua IH, Liu CJ, Tanwandee T, Gani R, Seto WK, Alam S, Young DY, Hamid S, Zheng MH, Kawaguchi T, Chan WK, Payawal D, Tan SS, Goh GBB, Strasser SI, Viet HD, Kao JH, Kim W, Kim SU, Keating SE, Yilmaz Y, Kamani L, Wang CC, Fouad Y, Abbas Z, Treeprasertsuk S, Thanapirom K, Al Mahtab M, Lkhagvaa U, Baatarkhuu O, Choudhury AK, Stedman CAM, Chowdhury A, Dokmeci AK, Wang FS, Lin HC, Huang JF, Howell J, Jia J, Alboraie M, Roberts SK, Yoneda M, Ghazinian H, Mirijanyan A, Nan Y, Lesmana CRA, Adams LA, Shiha G, Kumar M, Örmeci N, Wei L, Lau G, Omata M, Sarin SK, George J. The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic dysfunction-associated fatty liver disease. Hepatol Int 2025; 19:261-301. [PMID: 40016576 DOI: 10.1007/s12072-024-10774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/28/2024] [Indexed: 03/01/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) affects over one-fourth of the global adult population and is the leading cause of liver disease worldwide. To address this, the Asian Pacific Association for the Study of the Liver (APASL) has created clinical practice guidelines focused on MAFLD. The guidelines cover various aspects of the disease, such as its epidemiology, diagnosis, screening, assessment, and treatment. The guidelines aim to advance clinical practice, knowledge, and research on MAFLD, particularly in special groups. The guidelines are designed to advance clinical practice, to provide evidence-based recommendations to assist healthcare stakeholders in decision-making and to improve patient care and disease awareness. The guidelines take into account the burden of clinical management for the healthcare sector.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of MedicineSchool of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, Kaohsiung Medical University, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China
| | - Ian Homer Cua
- Institute of Digestive and Liver Diseases, St. Luke's Medical Center, Global City, Philippines
| | - Chun-Jen Liu
- Division of Gastroenterology and Hepatology, Department of Internal MedicineHepatitis Research CenterGraduate Institute of Clinical Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tawesak Tanwandee
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rino Gani
- Department of Internal Medicine, Hepatobiliary Division, Dr. Cipto Mangunkusumo National General Hospital, Universitas Indonesia, Pangeran Diponegoro Road No. 71St, Central Jakarta, 10430, Indonesia
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shahinul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Dan Yock Young
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Saeed Hamid
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Payawal
- Department of Medicine, Cardinal Santos Medical Center, Mandaluyong, Philippines
| | - Soek-Siam Tan
- Department of Hepatology, Selayang Hospital, Batu Caves, Malaysia
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Hang Dao Viet
- Internal Medicine Faculty, Hanoi Medical University, Hanoi, Vietnam
| | - Jia-Horng Kao
- Graduate Institute of Clinical MedicineDepartment of Internal MedicineHepatitis Research CenterDepartment of Medical Research, National Taiwan University College of Medicine, National Taiwan University, National Taiwan University Hospital, 1 Chang-Te Street, 10002, Taipei, Taiwan
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | - Chia-Chi Wang
- Buddhist Tzu Chi Medical Foundation and School of Medicine, Taipei Tzu Chi Hospital, Tzu Chi University, Taipei, Taiwan
| | - Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Cairo, Egypt
| | - Zaigham Abbas
- Department of Hepatogastroenterology, Dr.Ziauddin University Hospital, Clifton, Karachi, Pakistan
| | | | | | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Undram Lkhagvaa
- Department of Health Policy, School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Oidov Baatarkhuu
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Ashok Kumar Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, 110070, India
| | | | - Abhijit Chowdhury
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - A Kadir Dokmeci
- Department of Medicine, Ankara University School of Medicine, Ankara, Turkey
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Institute of Clinical Medicine, School of Medicine, Taipei Veterans General Hospital, National Yang-Ming Chiao Tung University, No. 201, Section 2, Shipai RdNo. 155, Section 2, Linong St, Beitou District, Taipei City, 112, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jess Howell
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Clayton, VIC, 3008, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, 3165, Australia
| | - Jidong Jia
- Liver Research Center, Beijing Key Laboratory of Translational Medicine On Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, 11884, Egypt
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Central Clinical School, The Alfred, Monash University, Melbourne, Australia
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hasmik Ghazinian
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Aram Mirijanyan
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Leon A Adams
- Medical School, Faculty of Medicine and Health Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Gamal Shiha
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansoura, Egypt
| | - Manoj Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Necati Örmeci
- Department of Gastroenterohepatology, Istanbul Health and Technology University, Istanbul, Turkey
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - George Lau
- Humanity and Health Medical Group, Humanity and Health Clinical Trial Center, Hong Kong SAR, China
- The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
- University of Tokyo, Tokyo, Japan
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
3
|
Uchinuma H, Matsushita M, Tanahashi M, Suganami H, Utsunomiya K, Kaku K, Tsuchiya K. Post-hoc analysis of the tofogliflozin post-marketing surveillance study (J-STEP/LT): Tofogliflozin improves liver function in type 2 diabetes patients regardless of BMI. J Diabetes Investig 2025; 16:615-628. [PMID: 39823131 PMCID: PMC11970296 DOI: 10.1111/jdi.14402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025] Open
Abstract
AIMS/INTRODUCTION Patients with type 2 diabetes are at high risk of developing steatotic liver disease (SLD). Weight loss has proven effective in treating metabolic dysfunction-associated steatotic liver disease (MASLD) in obese patients with type 2 diabetes, with sodium-glucose cotransporter 2 (SGLT2) inhibitors showing promising results. However, lean MASLD is more prevalent in Japan, necessitating alternative approaches to body weight reduction. MATERIALS AND METHODS We used the J-STEP/LT dataset including up to 3-year treatment data to analyze the effects of the SGLT2 inhibitor tofogliflozin on liver function and treatment safety and conducted a subgroup analysis based on body mass index (BMI; kg/m2, <20, 20-<23, 23-<25, 25-<30, and ≥30). RESULTS This study included 4,208 participants. Tofogliflozin significantly reduced alanine aminotransferase (ALT) levels in participants with baseline ALT levels >30 U/L across all BMI groups, with median changes of -12, -16, -13, -15, and -15 U/L, respectively (P = 0.9291 for trends). However, median changes in body weight with tofogliflozin were -2.00, -2.75, -2.00, -3.00, and -3.80 kg, respectively (P < 0.0001 for trends), with no significant weight loss observed in the BMI <20 group. ALT levels were also significantly decreased in participants who did not lose weight. Safety assessments according to BMI and age categories revealed no clear differences in the frequency of adverse events. CONCLUSIONS Tofogliflozin reduced ALT levels without substantial body weight reduction among lean participants. These findings suggest that SGLT2 inhibitors may be a viable treatment option for non-obese patients with type 2 diabetes and SLD.
Collapse
Affiliation(s)
- Hiroyuki Uchinuma
- Department of Diabetes and EndocrinologyUniversity of Yamanashi HospitalYamanashiJapan
| | | | | | | | | | - Kohei Kaku
- Division of Diabetes, Metabolism and EndocrinologyKawasaki Medical SchoolOkayamaJapan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and EndocrinologyUniversity of Yamanashi HospitalYamanashiJapan
| |
Collapse
|
4
|
Wang B, Yang Y, Yin Z, Yang W. The causal impact of body mass index on metabolic biomarkers and nonalcoholic fatty liver disease risk. Sci Rep 2025; 15:10314. [PMID: 40133380 PMCID: PMC11937590 DOI: 10.1038/s41598-024-84165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a growing global health concern linked to obesity. METHODS This study employed a Mendelian randomization approach to explore the causal influence of BMI on metabolic biomarkers and the subsequent risk of NAFLD. We analyzed data from multiple sources, including 249 metabolic traits, to establish direct and mediating relationships among BMI, metabolic factors, and NAFLD risk. RESULTS Our findings revealed a significant positive correlation between BMI and NAFLD across various datasets. We identified 176 metabolites associated with BMI, of which 106 were also linked to NAFLD. Importantly, 86 metabolites were found to mediate the relationship between BMI and NAFLD risk. Specifically, elevated levels of branched-chain amino acids, triglycerides, and certain cholesterol esters were notably associated with increased NAFLD risk, whereas changes in free cholesterol and phospholipid levels also played critical roles. CONCLUSION This study highlights the complex interactions between BMI, metabolic biomarkers, and NAFLD risk. By elucidating these relationships, we highlight potential targets for interventions aimed at reducing NAFLD incidence in populations with elevated BMI, ultimately contributing to improved metabolic health.
Collapse
Affiliation(s)
- Bo Wang
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yanjiang Yang
- Department of Rheumatology and Immunology, The People's Hospital of Qiandongnan Autonomous Prefecture, Kaili, 556000, Guizhou Province, China
| | - Zhaoqiang Yin
- Department of Minimally Invasive and Biliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China.
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
5
|
He S, Lv Y, Qiu J, Cui S, Gao Z, Peng L. Ta 4C 3 MXene Slows Progression of Fatty Liver Disease through Its Anti-Inflammatory and ROS-Scavenging Effects. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17217-17229. [PMID: 40051029 DOI: 10.1021/acsami.4c20945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Treating metabolic dysfunction-associated fatty liver disease (MAFLD) and reducing the occurrence of MAFLD-associated liver cancer remain challenging. Two-dimensional (2D) tantalum carbide (Ta4C3) MXene nanozymes (MXenzymes) exhibit antioxidant and anti-inflammatory activities and have thus attracted considerable attention in the fields of oncology and engineering. However, the potential mechanism of action and bioactive properties of Ta4C3 in MAFLD remain uncertain. In our study, Ta4C3 not only inhibited lipid accumulation and disrupted lipid metabolism in hepatocytes but also reduced cell death caused by fatty acids by decreasing intracellular reactive oxygen species (ROS) levels, which significantly promoted the polarization of M1 macrophages to M2 macrophages by alleviating oxidative stress and further suppressing inflammatory factor expression. In mice fed a methionine-choline-deficient (MCD) diet, Ta4C3 reduced lipid accumulation, the infiltration of inflammatory cells, and liver cell apoptosis by modulating the cellular microenvironment through its anti-inflammatory and antioxidant properties. Therefore, Ta4C3 can be used as a multifunctional bioactive material to alleviate hepatic steatosis and inflammation in individuals with MAFLD/metabolic dysfunction-associated steatohepatitis (MASH) because of its robust antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Shuying He
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yuerong Lv
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Jingnan Qiu
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Shudan Cui
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Zixian Gao
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Liang Peng
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
- Department of Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| |
Collapse
|
6
|
Song J, Chen S, Qian K, Ye W. Association of ultra-processed foods consumption with increased liver steatosis in U.S. adults. Front Nutr 2025; 12:1536989. [PMID: 40151346 PMCID: PMC11948534 DOI: 10.3389/fnut.2025.1536989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Background Recent studies demonstrated a strong association between dietary habits and liver health, particularly in the development of steatosis and fibrosis. This study aimed to examine the impact of ultra-processed foods (UPFs) on liver health, focusing specifically on their influence on the risks of liver steatosis and fibrosis. Methods A cross-sectional analysis was conducted on 4,992 participants aged 18 years and older from the 2017-2020 National Health and Nutrition Examination Survey (NHANES). Dietary intake was assessed using one or two 24-h dietary recalls, and foods were categorized by their processing level using the NOVA classification system. UPFs consumption was measured in grams and divided into quartiles. Liver health was assessed using controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) via elastography, to evaluate steatosis and fibrosis, respectively. Linear regression models were applied to assess the relationship between UPFs consumption and liver outcomes, adjusting for sociodemographic (age, sex, ethnicity), lifestyle (alcohol consumption, physical activity), and biomedical factors (liver enzyme levels). Results Higher UPF intake was significantly associated with increased CAP values, indicating a higher risk of liver steatosis. While liver fibrosis, measured by LSM, was also associated with UPF consumption, this relationship did not reach statistical significance. Multivariate analysis showed that increased UPF consumption did not significantly affect LSM (p = 0.110) but was strongly associated with elevated CAP values (p = 0.009). In participants with fatty liver (CAP > 248 dB/m), the association between UPF intake and CAP remained significant (p = 0.020). Participants in the highest quartile of UPFs consumption (Q4) exhibited higher CAP values compared to those in the lowest quartile (Q1) (β = 1.22; 95% CI: 1.02, 1.47). Stratified analysis revealed that the association between UPF intake and CAP was more pronounced in obese individuals (HR = 1.08, 95% CI: 1.03-1.15, p = 0.022) and those with high waist circumference (HR = 1.06, 95% CI: 1.01-1.10, p = 0.032). Conclusion These results underscore the adverse impact of UPFs on liver health, particularly by increasing steatosis, while the connection with fibrosis remains less straightforward.
Collapse
Affiliation(s)
- Jingru Song
- Department of Gastroenterology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Siqi Chen
- Department of Gastroenterology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kexin Qian
- Department of Gastroenterology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wei Ye
- Department of Gastroenterology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Fu W, Cheng GB, Zhao JL, Lv LY, Ding Y. Association of Life's Essential 8 and Life's Simple 7 with metabolic-associated fatty liver disease in the United States. World J Hepatol 2025; 17:97741. [PMID: 40027568 PMCID: PMC11866152 DOI: 10.4254/wjh.v17.i2.97741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/30/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Cardiovascular health (CVH) plays a crucial role in overall health, but its association with metabolic-associated fatty liver disease (MAFLD) remains unclear. AIM To investigate the relationship between CVH, measured using Life's Essential 8 (LE8) and Life's Simple 7 (LS7), and the prevalence of MAFLD. METHODS This cross-sectional study had a sample of 2234 individuals, representing approximately 120 million individuals in the United States. Baseline parameters were compared between the LE8 and LS7 groups. Logistic regression models were used to evaluate the relationship between LE8, LS7, and MAFLD, while taking into account confounding factors. The investigation employed restricted cubic splines to investigate non-linear associations. Subgroup analyses and sensitivity studies were performed to evaluate the strength and reliability of the results. RESULTS Higher LE8 and LS7 scores were significantly associated with a decreased risk of MAFLD, even after controlling for demographic, socioeconomic, and clinical variables. This association demonstrated a non-linear pattern, with the most dramatic risk reduction observed at higher CVH levels. Individual CVH components, notably healthy behaviors and factors, exhibited strong relationships with MAFLD. Subgroup analyses indicated consistent relationships across several demographics. Sensitivity tests utilizing other MAFLD definitions validated the robustness of the findings. CONCLUSION Higher adherence to CVH criteria, as indicated by LE8 and LS7 scores, is associated with a significantly lower risk of MAFLD. These results emphasize the need to advance CVH to control and avoid MAFLD.
Collapse
Affiliation(s)
- Wei Fu
- Department of Gastroenterology, The 925th of PLA Hospital, Guiyang 550009, Guizhou Province, China.
| | - Guo-Bin Cheng
- Department of Gastroenterology, 925 Hospital of PLA Joint Logistics Support Force, Guiyang 550009, Guizhou Province, China
| | - Jun-Long Zhao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Lin-Ya Lv
- Department of Gastroenterology, 925 Hospital of PLA Joint Logistics Support Force, Guiyang 550009, Guizhou Province, China
| | - Yao Ding
- Department of Gastroenterology, 925 Hospital of PLA Joint Logistics Support Force, Guiyang 550009, Guizhou Province, China
| |
Collapse
|
8
|
Zhu R, Xu C, Jiang S, Xia J, Wu B, Zhang S, Zhou J, Liu H, Li H, Lou J. Risk factor analysis and predictive model construction of lean MAFLD: a cross-sectional study of a health check-up population in China. Eur J Med Res 2025; 30:137. [PMID: 40001266 PMCID: PMC11863909 DOI: 10.1186/s40001-025-02373-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
AIM Cardiovascular disease morbidity and mortality rates are high in patients with metabolic dysfunction-associated fatty liver disease (MAFLD). The objective of this study was to analyze the risk factors and differences between lean MAFLD and overweight MAFLD, and establish and validate a nomogram model for predicting lean MAFLD. METHODS This retrospective cross-sectional study included 4363 participants who underwent annual health checkup at Yuyao from 2019 to 2022. The study population was stratified into three groups: non-MAFLD, lean MAFLD (defined as the presence of fatty liver changes as determined by ultrasound in individuals with a BMI < 25 kg/m2), and overweight MAFLD (BMI ≥ 25.0 kg/m2). Subsequent modeling analysis was conducted in a population that included healthy subjects with < 25 kg/m2 (n = 2104) and subjects with lean MAFLD (n = 849). The study population was randomly split (7:3 ratio) to a training vs. a validation cohort. Risk factors for lean MAFLD was identify by multivariate regression of the training cohort, and used to construct a nomogram to estimate the probability of lean MAFLD. Model performance was examined using the receiver operating characteristic (ROC) curve analysis and k-fold cross-validation (k = 5). Decision curve analysis (DCA) was applied to evaluate the clinical usefulness of the prediction model. RESULTS The multivariate regression analysis indicated that the triglycerides and glucose index (TyG) was the most significant risk factor for lean MAFLD (OR: 4.03, 95% CI 2.806-5.786). The restricted cubic spline curves (RCS) regression model demonstrated that the relationships between systolic pressure (SBP), alanine aminotransferase (ALT), serum urate (UA), total cholesterol (TCHO), triglyceride (TG), triglyceride glucose (TyG) index, high density lipoprotein cholesterol (HDLC), and MAFLD were nonlinear and the cutoff values for lean MAFLD and overweight MAFLD were different. The nomogram was constructed based on seven predictors: glycosylated hemoglobin A1c (HbA1c), serum ferritin (SF), ALT, UA, BMI, TyG index, and age. In the validation cohort, the area under the ROC curve was 0.866 (95% CI 0.842-0.891), with 83.8% sensitivity and 76.6% specificity at the optimal cutoff. The PPV and NPV was 63.3% and 90.8%, respectively. Furthermore, we used fivefold cross-validation and the average area under the ROC curve was 0.866 (Figure S3). The calibration curves for the model's predictions and the actual outcomes were in good agreement. The DCA findings demonstrated that the nomogram model was clinically useful throughout a broad threshold probability range. CONCLUSIONS Lean and overweight MAFLD exhibit distinct metabolic profiles. The nomogram model developed in this study is designed to assist clinicians in the early identification of high-risk individuals with lean MAFLD, including those with a normal BMI but at metabolic risk, as well as those with abnormal blood lipid, glucose, uric acid or transaminase levels. In addition, this model enhances screening efforts in communities and medical screening centers, ultimately ensuring more timely and effective medical services for patients.
Collapse
Affiliation(s)
- Ruya Zhu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Caicai Xu
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China
| | - Suwen Jiang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jianping Xia
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Boming Wu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Sijia Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongliang Liu
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, Zhejiang, China.
| | - Jianjun Lou
- Chronic Liver Disease Center, The Affiliated Yangming Hospital of Ningbo University, Zhejiang, 315400, China.
| |
Collapse
|
9
|
Lu Y, Li X, Ma S, Ding M, Yang F, Pang X, Sun J, Li X. Broccoli ( Brassica oleracea L. var. italica Planch) alleviates metabolic-associated fatty liver disease through regulating gut flora and lipid metabolism via the FXR/LXR signaling pathway. Food Funct 2025; 16:1218-1240. [PMID: 39903517 DOI: 10.1039/d4fo03731f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The increased consumption of dietary fats contributes to the development of MAFLD (metabolic fatty liver disease). The ability of broccoli to enhance lipid metabolism has attracted researchers' attention. Researchers fed C57BL/6 mice a 12-week HFD to ensure the induction of MAFLD. The findings indicated that broccoli floret juice could effectively relieve MAFLD. Broccoli is helpful for reducing weight, blood glucose levels, fat accumulation, and insulin resistance associated with MAFLD and reduces the concentrations of TC, TG, LDL-C, GOT, GPT, IL-1β, IL-6, CCL4, and MCP1. Broccoli can increase the concentration of HDL-C, CAT, GSH-Px, SOD, and T-AOC, relieve inflammation and hepatic and ileum damage, and improve the antioxidant capacity of the body. Also, broccoli can optimize the structure of intestinal flora, promote the growth of Allobaculum, Muribaculaceae, Akkermansia, Eubacterium, and Bacteroides, and reduce bile acid deposition. In addition, the FXR/LXRα signaling system is impacted by broccoli, which is capable of raising the average levels of expression of the Fxr, SHP, and Cyp7a1 genes and proteins and reducing those of the genes for Fasn, Lpin 1, Dgat 2, Scd1, LXRα, and SREBP-1c.
Collapse
Affiliation(s)
- Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Xin Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Shaotong Ma
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Meng Ding
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Feiyu Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| |
Collapse
|
10
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-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: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
11
|
Wu X, Pan T, Fang Z, Hui T, Yu X, Liu C, Guo Z, Liu C. Identification of EGR1 as a Key Diagnostic Biomarker in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Through Machine Learning and Immune Analysis. J Inflamm Res 2025; 18:1639-1656. [PMID: 39925925 PMCID: PMC11806694 DOI: 10.2147/jir.s499396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/25/2025] [Indexed: 02/11/2025] Open
Abstract
Background Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), as a common chronic liver condition globally, is experiencing an increasing incidence rate which poses significant health risks. Despite this, the detailed mechanisms underlying the disease's onset and progression remain poorly understood. In this study, we aim to identify effective diagnostic biomarkers for MASLD using microarray data combined with machine learning techniques, which will aid in further understanding the pathogenesis of MASLD. Methods We collected six datasets from the Gene Expression Omnibus (GEO) database, using five of them as training sets and one as a validation set. We employed three machine learning methods-LASSO, SVM, and Random Forest (RF)-to identify hub genes associated with MASLD. These genes were further validated using the external dataset GSE164760. Additionally, functional enrichment analysis, immune infiltration analysis, and immune function analysis were conducted. A TF-miRNA-mRNA network was constructed, and single-cell RNA sequencing was used to determine the distribution of key genes within key cell clusters. Finally, the expression of the key genes was further validated using the palmitic acid-induced AML-12 cell line and the MCD mouse model. Results In this study, through differential gene expression (DEGs) analysis and machine learning techniques, we successfully identified 10 hub genes. Among these, the key gene EGR1 was validated and screened using an external dataset, with an area under the curve (AUC) of 0.882. Enrichment analyses and immune infiltration assessments revealed multiple pathways involving EGR1 in the pathogenesis and progression of MASLD, showing significant correlations with various immune cells. Furthermore, additional cellular experiments and animal model validations confirmed that the expression trends of EGR1 are highly consistent with our analytical findings. Conclusion Our research has confirmed EGR1 as a key gene in MASLD, providing novel insights into the disease's pathogenesis and identifying new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Xuanlin Wu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tao Pan
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Titi Hui
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaoxiao Yu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zihao Guo
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
12
|
Bae JH, Cho YM. Incretin hormones: Revolutionizing the treatment landscape for kidney and liver diseases in type 2 diabetes and obesity. J Diabetes Investig 2025; 16:183-186. [PMID: 39460581 PMCID: PMC11786174 DOI: 10.1111/jdi.14336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Several ongoing trials are evaluating incretin-based therapies, including GLP-1 receptor agonists, for their effects on CKD and MASLD. These studies will offer insights into their potential for metabolic diseases in people with type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Jae Hyun Bae
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
| | - Young Min Cho
- Department of Internal MedicineSeoul National University HospitalSeoulKorea
- Department of Internal MedicineSeoul National University College of MedicineSeoulKorea
| |
Collapse
|
13
|
Ye Z, Xie E, Guo Z, Gao Y, Han Z, Dou K, Zheng J. Association of Liver Fibrosis Markers with Mortality Outcomes in Patients with Chronic Kidney Disease and Coronary Artery Disease: Insights from the NHANES 1999-2018 Data. Cardiorenal Med 2025; 15:153-163. [PMID: 39837280 PMCID: PMC11844702 DOI: 10.1159/000543500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
INTRODUCTION The objective of this research was to explore the possible link between markers of liver fibrosis and survival rates in a group of adults who have been diagnosed with both chronic kidney disease (CKD) and coronary artery disease (CAD). METHODS The National Health and Nutrition Examination Survey (NHANES) data (1999-2018) for participants with both CAD and CKD were analyzed. The fibrosis-4 index (FIB-4), Nonalcoholic Fatty Liver Score (NFS), Forns index, and aspartate aminotransferase/alanine aminotransferase (AST/ALT) ratio were identified as crucial biomarkers. All-cause and cardiovascular disease (CVD) mortality were primary outcomes, assessed using Cox models, Kaplan-Meier curves, and receiver operating characteristic (ROC) analysis. RESULTS A total of 1,192 CKD and CAD patients were included. The Cox regression analysis revealed substantial correlations between elevated FIB-4, NFS, Forns index, and AST/ALT levels and a heightened risk of all-cause (hazard ratio [HR]: 1.188, 95% confidence interval [CI]: 1.108-1.274; HR: 1.145, 95% CI: 1.069-1.227; HR: 1.142, 95% CI: 1.081-1.201; HR: 1.316, 95% CI: 1.056-1.639, respectively) and CVD mortality (HR: 1.133, 95% CI: 1.007-1.275; HR: 1.155, 95% CI: 1.024-1.303; HR: 1.208, 95% CI: 1.109-1.316 and HR: 1.636, 95% CI: 1.203-2.224, respectively). The ROC analysis indicated comparable predictive accuracy for all three biomarkers, with AST/ALT showing slightly superior performance. CONCLUSION Liver fibrosis markers, including AST/ALT, NFS, Forns index and FIB-4, are significant mortality predictors in CAD-CKD patients. The AST/ALT ratio, being easily measurable, may serve as an effective predictive tool for risk stratification in this population.
Collapse
Affiliation(s)
- Zixiang Ye
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Enmin Xie
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziyu Guo
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Zhongwei Han
- Department of Cardiology, Yantai Muping District Hospital of Traditional Chinese Medicine, Yantai, China
| | - Kefei Dou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
14
|
Seyedi A, Rabizadeh S, Reyhan SK, Hobaby S, Afshari SA, Shomalzadeh M, Nabipoorashrafi SA, Soran NA, Yadegar A, Mohammadi F, Moosaei F, Saffari E, Riazi S, Sarv F, Nakhjavani M, Pazouki A, Esteghamati A. Impact of bariatric surgery on liver fibrosis indices among type 2 diabetes patients in a national cohort. Sci Rep 2025; 15:1235. [PMID: 39775004 PMCID: PMC11707195 DOI: 10.1038/s41598-025-85427-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Obesity is related to liver fibrosis, a condition marked by the collection of scar tissue in the liver due to the development of a profibrotic environment, which includes increased hepatocellular death and elevated reactive oxygen species production. The aim of study is to evaluate the effect of bariatric surgery on the association between liver fibrosis indices and obesity. This is a retrospective cohort, evaluating 1205 individuals diagnosed with type 2 diabetes (T2D) and living with obesity, who experienced bariatric surgery. These patients living with T2D and obesity were monitored after bariatric surgery for two years. The trajectory of biochemical markers and liver fibrosis indices were evaluated at five visits. These liver indices were Fibrosis-4 (FIB-4) index, aspartate aminotransferase (AST) to platelet ratio index, and non-alcoholic fatty liver disease (NAFLD) fibrosis score. FIB-4 index demonstrated notable trends based on its values. It showed an initial increase observed at the three-months visit, followed by a decline up to one year with a slight increase at the last follow-up (P-trend < 0.001). It should be mentioned that, mean FIB-4 in patients with FIB-4 ≥ 1.3 (pre-operation) did not exceed the value of 2.00, which is lower than the cut-off value of high risk for liver cirrhosis (FIB-4 ≥ 2.67). In addition, the NAFLD fibrosis score (NFS) demonstrated a substantial decline from - 0.32 ± 1.32 pre-operation to -0.86 ± 1.15 at the two-year mark (P-trend < 0.001). Finally, the AST to platelet ratio index (APRI) decreased from 0.27 ± 0.20 pre-operation to 0.23 ± 0.12 at the 12-month follow-up. Bariatric surgery significantly improves NFS and cause alterations in APRI and Fib-4 index levels without increasing the risk of liver cirrhosis development among patients with T2D and obesity.
Collapse
Affiliation(s)
- Arsalan Seyedi
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Soghra Rabizadeh
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Sahar Karimpour Reyhan
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran.
| | - Sara Hobaby
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Amin Afshari
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Mostafa Shomalzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Nabipoorashrafi
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Nasrin Asgari Soran
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasoule-Akram Hospital, Mansouri st., Niyayesh Ave., Sattarkhan St., Tehran, Iran
| | - Amirhossein Yadegar
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Fatemeh Mohammadi
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Fatemeh Moosaei
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
- International Surgical Research Association (ISRA), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, P.O. Box 13145784, Tehran, Iran
| | - Elahe Saffari
- Isfahan University of Medical science, Isfahan, Iran
| | - Sajedeh Riazi
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasoule-Akram Hospital, Mansouri st., Niyayesh Ave., Sattarkhan St., Tehran, Iran
| | - Fatemeh Sarv
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasoule-Akram Hospital, Mansouri st., Niyayesh Ave., Sattarkhan St., Tehran, Iran
| | - Manouchehr Nakhjavani
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran
| | - Abdolreza Pazouki
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Rasoule-Akram Hospital, Mansouri st., Niyayesh Ave., Sattarkhan St., Tehran, Iran
| | - Alireza Esteghamati
- Endocrinology and Metabolism Research Center (EMRC), School of Medicine, Vali-Asr Hospital, Imam Khomeini Hospital, Tehran, Iran.
| |
Collapse
|
15
|
Sakamoto K, Butera MA, Zhou C, Maurizi G, Chen B, Ling L, Shawkat A, Patlolla L, Thakker K, Calle V, Morgan DA, Rahmouni K, Schwartz GJ, Tahiri A, Buettner C. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab 2025; 37:121-137.e6. [PMID: 39437790 PMCID: PMC11711004 DOI: 10.1016/j.cmet.2024.09.012] [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: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The mechanisms underlying obesity-induced insulin resistance remain incompletely understood, as impaired cellular insulin signaling, traditionally considered the primary driver of insulin resistance, does not always accompany impaired insulin action. Overnutrition rapidly increases plasma norepinephrine (NE), suggesting overactivation of the sympathetic nervous system (SNS). However, the role of the SNS in obesity is controversial, as both increased and decreased SNS activity (SNA) have been reported. Here, we show that reducing catecholamine (CA) release from the SNS protects against overnutrition-induced insulin resistance as well as hyperglucagonemia, adipose tissue dysfunction, and fatty liver disease, as we demonstrate utilizing a mouse model of inducible and peripherally restricted deletion of tyrosine hydroxylase (th; THΔper). A key mechanism through which heightened SNA induces insulin resistance is by triggering adipose tissue lipolysis. Increased SNA emerges as a critical driver in the pathogenesis of overnutrition-induced insulin resistance and metabolic disease independent of cellular insulin signaling.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary A Butera
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxue Zhou
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Maurizi
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bandy Chen
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Ling
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adham Shawkat
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Likhitha Patlolla
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kavira Thakker
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Victor Calle
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gary J Schwartz
- Department of Medicine & Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Azeddine Tahiri
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
Yang X, Rao H, Yuan Y, Hu N, Zhang X, Zeng Y, Xia G. Correlation analysis of the triglyceride-glucose index and related parameters in metabolic dysfunction-associated fatty liver disease. Sci Rep 2025; 15:23. [PMID: 39748005 PMCID: PMC11695697 DOI: 10.1038/s41598-024-84809-y] [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: 11/10/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
This study aimed to investigate the correlation and predictive value of TyG and related parameters with metabolic dysfunction-associated fatty liver disease (MAFLD) MAFLD. This study retrospectively included individuals who underwent health examinations and abdominal ultrasound from July 2021 to June 2024 at the Affiliated Hospital of Southwest Medical University, Sichuan Province, China. A total of 71,299 subjects' clinical and laboratory data were extracted, the correlation between TyG and related parameters and MAFLD was analyzed via univariate and multivariate logistic regression methods, and the nonlinear relationship between the TyG index and the risk of MAFLD was explored via restricted cubic spline (RCS) analysis. The predictive value of TyG and related parameters for MAFLD was assessed using the receiver operating characteristic (ROC) curves and the area under the ROC curve (AUC). TyG and related parameters were positively correlated with MAFLD, and the results remained unchanged after adjustment for the corresponding parameters. RCS analysis revealed a significant dose‒response relationship between TyG and related parameters and MAFLD. ROC curve analysis revealed AUC values of 0.83 (0.82-0.83), 0.92 (0.91-0.92), 0.90 (0.90-0.91), and 0.87 (0.87-0.88) for TyG, TyG-BMI, TyG-WC, and TyG-WHR, respectively. Subgroup analyses revealed that the TyG index and related parameters had greater predictive value in the female, younger, and BMI < 23.7 populations.
Collapse
Affiliation(s)
- Xin Yang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Street Taiping No.25, Region Jiangyang, Luzhou, 646099, Sichuan, China
| | - Huiting Rao
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Street Taiping No.25, Region Jiangyang, Luzhou, 646099, Sichuan, China
| | - Yi Yuan
- Department of Presbyatrics, Luzhou People's Hospital, Luzhou, China
| | - Nan Hu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Street Taiping No.25, Region Jiangyang, Luzhou, 646099, Sichuan, China
| | - Xinmei Zhang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Street Taiping No.25, Region Jiangyang, Luzhou, 646099, Sichuan, China
| | - Yingxin Zeng
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Street Taiping No.25, Region Jiangyang, Luzhou, 646099, Sichuan, China
| | - Guodong Xia
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Street Taiping No.25, Region Jiangyang, Luzhou, 646099, Sichuan, China.
- Department of Health Management Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
17
|
Sotoudeheian M. Value of Mac-2 Binding Protein Glycosylation Isomer (M2BPGi) in Assessing Liver Fibrosis in Metabolic Dysfunction-Associated Liver Disease: A Comprehensive Review of its Serum Biomarker Role. Curr Protein Pept Sci 2025; 26:6-21. [PMID: 38982921 DOI: 10.2174/0113892037315931240618085529] [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: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 07/11/2024]
Abstract
Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) is a broad condition characterized by lipid accumulation in the liver tissue, which can progress to fibrosis and cirrhosis if left untreated. Traditionally, liver biopsy is the gold standard for evaluating fibrosis. However, non-invasive biomarkers of liver fibrosis are developed to assess the fibrosis without the risk of biopsy complications. Novel serum biomarkers have emerged as a promising tool for non-invasive assessment of liver fibrosis in MAFLD patients. Several studies have shown that elevated levels of Mac-2 binding protein glycosylation isomer (M2BPGi) are associated with increased liver fibrosis severity in MAFLD patients. This suggests that M2BPGi could serve as a reliable marker for identifying individuals at higher risk of disease progression. Furthermore, the use of M2BPGi offers a non-invasive alternative to liver biopsy, which is invasive and prone to sampling errors. Overall, the usage of M2BPGi in assessing liver fibrosis in MAFLD holds great promise for improving risk stratification and monitoring disease progression in affected individuals. Further research is needed to validate its utility in clinical practice and establish standardized protocols for its implementation.
Collapse
|
18
|
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
|
19
|
Keles U, Kalem-Yapar NE, Hultén H, Zhao LN, Kaldis P. Impact of Short-Term Lipid Overload on Whole-Body Physiology. Mol Cell Biol 2024; 45:47-58. [PMID: 39726368 DOI: 10.1080/10985549.2024.2438814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Complex metabolic diseases due to overnutrition such as obesity, type 2 diabetes, and fatty liver disease are a major burden on the healthcare system worldwide. Current research primarily focuses on disease endpoints and trying to understand underlying mechanisms at relatively late stages of the diseases, when irreversible damage is already done. However, complex interactions between physiological systems during disease development create a problem regarding how to build cause-and-effect relationships. Therefore, it is essential to understand the early pathophysiological effects of overnutrition, which can help us understand the origin of the disease and to design better treatment strategies. Here, we focus on early metabolic events in response to high-fat diets (HFD) in rodents. Interestingly, insulin resistance, fatty liver, and obesity-promoting systemic inflammatory responses are evident within a week when mice are given consecutive HFD meals. However, as shown in human studies, these effects are usually not visible after a single meal. Overall, these results suggest that sustained HFD-intake within days can create a hyperlipidemic environment, globally remodeling metabolism in all affected organs and resembling some of the important disease features.
Collapse
Affiliation(s)
- Umur Keles
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Nisan Ece Kalem-Yapar
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Hanna Hultén
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Li Na Zhao
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Clinical Research Centre (CRC), Malmö, Sweden
| |
Collapse
|
20
|
Lu CW, Chou TJ, Wu TY, Lee YH, Yang HJ, Huang KC. PNPLA3 and SAMM50 variants are associated with lean nonalcoholic fatty liver disease in Asian population. Ann Hepatol 2024; 30:101761. [PMID: 39638042 DOI: 10.1016/j.aohep.2024.101761] [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: 03/05/2024] [Revised: 08/02/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION AND OBJECTIVES Lean adults with nonalcoholic fatty liver disease (NAFLD) have a higher risk of metabolic syndrome than lean controls. The study aimed to investigate the clinical and genetic features of lean NAFLD which remain unclear in Asian populations. MATERIALS AND METHODS This was a genetic cohort study conducted in the HAVO Health Exam Clinic in 2020-2021 in Taiwan. Adults with a body mass index less than 24 kg/m2 were enrolled. Fatty liver was defined by ultrasonography. The candidate gene approach was based on the library of the NHGRI-EBI website. After removing duplication and nonsignificant variants, rs738409 in the PNPLA3 gene and rs3761472 in the SAMM50 gene were chosen. Multiple logistic regression models and receiver operating characteristic (ROC) curves were used. RESULTS A total of 1652 lean controls and 602 lean NAFLD patients were enrolled. The average age was 43.8 ± 11.5 years. Lean NAFLD subjects were older and had a higher percentage of metabolic syndrome (case vs. control: 10.5 % vs. 1.5 %). The GG genotypes of PNPLA3 rs12483959 (OR: 3.06; 95% CI: 2.15-4.37) and SAMM50 rs3761472 (OR: 2.90; 95% CI: 2.04-4.14) had a higher risk of fatty liver after adjusting for BMI and metabolic syndrome. The areas under the ROC curve for PNPLA3 rs738409 and SAMM50 rs3761472 in the detection of lean NAFLD were 0.859 (95%CI: 0.841, 0.877) and 0.860 (95%CI: 0.843, 0.877), respectively. CONCLUSIONS PNPLA3 rs738409 and SAMM50 rs3761472 gene polymorphisms are associated with a higher risk of fatty liver in lean individuals independent of BMI and metabolic syndrome in Asian populations.
Collapse
Affiliation(s)
- Chia-Wen Lu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Jung Chou
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Family Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Tsan-Yu Wu
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Jen Yang
- Min-Sheng General Hospital, Taoyuan, Taiwan; Share Hope Medicine Co., Ltd., Taoyuan, Taiwan
| | - Kuo-Chin Huang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Family Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan.
| |
Collapse
|
21
|
Kim HK, Kim DY, Kang S, Kim H, Kim JM, Go GW. Lean metabolic dysfunction-associated steatotic disease is reversed by betulinic acid, a therapeutic triterpene from birch bark. FOOD BIOSCI 2024; 62:104376. [DOI: 10.1016/j.fbio.2024.104376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
22
|
Yuan HY, Liu WY, Feng G, Chen SD, Jin XZ, Chen LL, Song ZJ, Li K, Byrne CD, Targher G, Tian N, Li G, Zhang XL, George J, Zhou M, Wang F, Zheng MH. Associations between cuprotosis-related genes and the spectrum of metabolic dysfunction-associated fatty liver disease: An exploratory study. Diabetes Obes Metab 2024; 26:5757-5775. [PMID: 39285685 DOI: 10.1111/dom.15946] [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: 06/22/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024]
Abstract
AIMS To explore the associations between cuprotosis-related genes (CRGs) across different stages of liver disease in metabolic dysfunction-associated fatty liver disease (MAFLD), including hepatocellular carcinoma (HCC). MATERIALS AND METHODS We analysed several bulk RNA sequencing datasets from patients with MAFLD (n = 331) and MAFLD-related HCC (n = 271) and two MAFLD single-cell RNA sequencing datasets. To investigate the associations between CRGs and MAFLD, we performed differential correlation, logistic regression and functional enrichment analyses. We also validated the findings in an independent Wenzhou PERSONS cohort of MAFLD patients (n = 656) used for a genome-wide association study (GWAS). RESULTS GLS, GCSH and ATP7B genes showed significant differences across the MAFLD spectrum and were significantly associated with liver fibrosis stages. GLS was closely associated with fibrosis stages in patients with MAFLD and those with MAFLD-related HCC. GLS is predominantly expressed in monocytes and T cells in MAFLD. During the progression of metabolic dysfunction-associated fatty liver to metabolic-associated steatohepatitis, GLS expression in T cells decreased. GWAS revealed that multiple single nucleotide polymorphisms in GLS were associated with clinical indicators of MAFLD. CONCLUSIONS GLS may contribute to liver inflammation and fibrosis in MAFLD mainly through cuprotosis and T-cell activation, promoting the progression of MAFLD to HCC. These findings suggest that cuprotosis may play a role in MAFLD progression, potentially providing new insights into MAFLD pathogenesis.
Collapse
Affiliation(s)
- Hai-Yang Yuan
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gong Feng
- Xi'an Medical University, Xi'an, China
| | - Sui-Dan Chen
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin-Zhe Jin
- Department of Laboratory Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li-Li Chen
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zi-Jun Song
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Li
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Na Tian
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gang Li
- Department of Infectious, Jining No.1 People's Hospital, Jining, China
| | - Xin-Lei Zhang
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jacob George
- Storr Liver Centre, Westmead Hospital and University of Sydney, Westmead, Australia
| | - Meng Zhou
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| |
Collapse
|
23
|
Sun W, Lv Y, Wang L, Yu H, Yi H, Wang Y, Han J, Liu Y, Miao C, Li J, Zhang Y, Wang M, Chen L, Pandol SJ, Li L. Comparison of Risk Factors Between Lean and Nonlean Metabolic Dysfunction-Associated Steatotic Liver Disease in Individuals With Type 2 Diabetes: A Multicenter Study. Endocr Pract 2024; 30:1171-1179. [PMID: 39332499 DOI: 10.1016/j.eprac.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE A multicenter study in patients with type 2 diabetes mellitus (T2DM) was performed to assess the differences of liver steatosis and fibrosis between lean and nonlean individuals. METHODS Patients with T2DM from 16 centers were recruited and underwent transient elastography examination for diagnosis of liver steatosis and fibrosis. Clinical information, such as diabetes status, serum lipids profiles, and inflammatory markers, were collected. Potential risk factors of liver steatosis and fibrosis in lean (body mass index [BMI] < 23 kg/m2) and nonlean (BMI ≥ 23 kg/m2) groups were analyzed. RESULTS A total of 1762 patients were included. The prevalence of liver steatosis and fibrosis in the lean group was 44.7% and 23.4%, respectively. The prevalence of hypertension and cardiovascular disease was higher in lean patients when compared with nonlean group. Lean patients with liver steatosis or fibrosis were older, had longer diabetes duration, lower levels of homeostatic model assessment for insulin resistance and serum lipids. The BMI, visceral fat area, and triglyceride were among the most significant correlators of liver steatosis for both nonlean and lean patients. However, lipid profiles were different between the two groups. Besides, insulin resistance, BMI, and lipid levels were not observed to be associated with fibrosis in the lean group. CONCLUSION In lean patients with T2DM, liver steatosis and fibrosis were less associated with insulin resistance. Risk factors of liver steatosis were different between lean and nonlean patients, indicating the necessity of risk stratification and tailored management strategies.
Collapse
Affiliation(s)
- Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Yingqi Lv
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Li Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China; Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Hekai Yu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - He Yi
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Yifan Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China
| | - Jing Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yingzhao Liu
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Congqing Miao
- Department of Endocrinology, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Jie Li
- Department of Endocrinology, Nanjing Central Hospital, Nanjing, China
| | - Yan Zhang
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Mengying Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lei Chen
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Stephen J Pandol
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California; Basic and Translational Pancreatic Research, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Southeast University, Nanjing, China; Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China.
| |
Collapse
|
24
|
Babu AF, Palomurto S, Kärjä V, Käkelä P, Lehtonen M, Hanhineva K, Pihlajamäki J, Männistö V. Metabolic signatures of metabolic dysfunction-associated steatotic liver disease in severely obese patients. Dig Liver Dis 2024; 56:2103-2110. [PMID: 38825414 DOI: 10.1016/j.dld.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024]
Abstract
BACKROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) can lead to liver fibrosis, cirrhosis, and hepatocellular carcinoma. Still, most patients with MASLD die from cardiovascular diseases indicating metabolic alterations related to both liver and cardiovascular pathology. AIMS AND METHODS The aim of this study was to assess biologic pathways behind MASLD progression from steatosis to metabolic dysfunction-associated steatohepatitis (MASH) using non-targeted liquid chromatography-mass spectrometry analysis in 106 severely obese individuals (78 women, mean age 47.7 7 ± 9.2 years, body mass index 41.8 ± 4.3 kg/m²) undergoing laparoscopic Roux-en-Y gastric bypass. RESULTS We identified several metabolites that are associated with MASLD progression. Most importantly, we observed a decrease of lysophosphatidylcholines LPC(18:2), LPC(18:3), and LPC(20:3) and increase of xanthine when comparing those with steatosis to those with MASH. We found that indole propionic acid and threonine were negatively correlated to fibrosis, but not with the metabolic disturbances associated with cardiovascular risk. Xanthine, ketoleucine, and tryptophan were positively correlated to lobular inflammation and ballooning but also with insulin resistance, and dyslipidemia, respectively. The results did not change when taking into account the most important genetic risk factors of MASLD. CONCLUSIONS Our findings suggest that there are several separate biological pathways, some of them independent of insulin resistance and dyslipidemia, associating with MASLD.
Collapse
Affiliation(s)
- Ambrin Farizah Babu
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland
| | - Saana Palomurto
- Department of Surgery, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Pirjo Käkelä
- Department of Surgery, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, Faculty of Health Science, University of Eastern Finland, 70211 Kuopio, Finland; LC-MS Metabolomics Center, Biocenter Kuopio, 70211 Kuopio, Finland
| | - Kati Hanhineva
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, 20014 Turku, Finland
| | - Jussi Pihlajamäki
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70210 Kuopio Finland
| | - Ville Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland.
| |
Collapse
|
25
|
Suoh M, Esmaili S, Eslam M, George J. Metabolic (dysfunction)-associated fatty liver disease metrics and contributions to liver research. Hepatol Int 2024; 18:1740-1755. [PMID: 39412611 PMCID: PMC11632019 DOI: 10.1007/s12072-024-10731-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/06/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND The international consensus to revise non-alcoholic fatty liver disease to metabolic (dysfunction)-associated fatty liver disease (MAFLD) in 2020 attracted significant attention. The impact of the MAFLD definition on the research community has not been objectively assessed. We conducted an analysis of systematically collected literature on MAFLD to understand its research impact. METHODS From PubMed, Web of Science, and Scopus, the literature adopting MAFLD, written in English, and published from 2020 to 10 October 2023 was collected. The publication metrics, including publication counts, publishing journals, author countries, author keywords, and citation information, were analyzed to evaluate the research impact and key topics on MAFLD. RESULTS 1469 MAFLD-related papers were published in 434 journals with a steady increase in the number. The intense publishing and citations activity on MAFLD indicates the large impact of the redefinition. Topic assessment with keyword and citation analysis revealed a transition from the proposal and discussion of the redefinition to clinical characterization of MAFLD with a focus on metabolic dysfunction. Moreover, the diagnostic criteria for MAFLD showed better performance in predicting hepatic and extrahepatic outcomes compared to NAFLD. The publications were from 99 countries with evidence of strong regional and global collaboration. Multiple international societies and stakeholders have endorsed MAFLD for its utility in clinical practice, improving patient management and promoting multidisciplinary care, while alleviating stigma. CONCLUSION This survey provides a quantitative measure of the considerable international impact and contributions of the MAFLD definition towards liver research and as part of the spectrum of cardiometabolic disorders.
Collapse
Affiliation(s)
- Maito Suoh
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, 176 Hawkesbury Rd, Westmead, NSW, 2145, Australia
| | - Saeed Esmaili
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, 176 Hawkesbury Rd, Westmead, NSW, 2145, Australia
| | - Mohammed Eslam
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, 176 Hawkesbury Rd, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, 176 Hawkesbury Rd, Westmead, NSW, 2145, Australia.
| |
Collapse
|
26
|
Jiang M, Pan Z, George J, Eslam M. Clinical features and mortality outcomes of patients with MASLD only compared to those with MAFLD and MASLD. Hepatol Int 2024; 18:1731-1739. [PMID: 39196505 DOI: 10.1007/s12072-024-10721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND AND AIMS The applicability of the proposed metabolic dysfunction-associated steatotic liver disease (MASLD) definition has not been validated. We aimed to characterize the profiles and long-term survival of people meeting the criteria for MASLD, but not that of metabolic dysfunction-associated fatty liver disease (MAFLD), i.e. MASLD only. METHODS Using data from the Third National Health and Nutrition Examination Survey (NHANES III) 1988-1994, 7791 adult participants were included and categorized into four distinct groups: no SLD, non-MAFLD MASLD, MASLD-MAFLD, and cryptogenic SLD (steatosis without metabolic dysfunction). RESULTS Participants in the MASLD-only group were younger and had better metabolic profiles and fibrosis degree compared to those with MASLD-MAFLD and those with no SLD. Their profiles were comparable to those with cryptogenic SLD. Similarly, the MASLD-only group tend to have lower cumulative incidence of all-cause and cardiovascular mortality. Clustering analysis showed that MASLD only clusters differently from individuals with MASLD-MAFLD. CONCLUSIONS MASLD only is a distinct clinical group with substantial heterogeneity compared to those captured using the MAFLD criteria.
Collapse
Affiliation(s)
- Mingqian Jiang
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
- Department of Endocrinology and Metabolism, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
27
|
Huang N, Su X, Yu T, Wu X, Lu B, Sun W, Yao L, Wang M, Wang Y, Wu W, Liu Y, Yang T, Gao R, Miao C, Li L. Serum 25-hydroxy vitamin D level is associated with elastography-detected liver fibrosis in patients with type 2 diabetes mellitus in China. Front Endocrinol (Lausanne) 2024; 15:1420088. [PMID: 39698035 PMCID: PMC11653015 DOI: 10.3389/fendo.2024.1420088] [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: 04/19/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024] Open
Abstract
Objective In this cross-sectional study including patients with type 2 diabetes mellitus (T2DM) we aimed to explore the relationship between serum 25-hydroxy vitamin (25(OH)D) level and liver steatosis and fibrosis in the Chinese population. Methods Patients visiting 16 clinical centers with T2DM were recruited. Their liver steatosis and fibrosis status were then assessed using elastography. Factors associated with steatosis and fibrosis were explored using regression analysis. Correlations between serum 25(OH)D levels and other patient characteristics were analyzed using linear regression. Results In total, 1,513 patients with T2DM were included in the study. The prevalence of steatosis and fibrosis was 69.7%, and 34.6%, separately. A lower level of 25(OH)D was detected in patients with liver steatosis compared to those without, although it was not an independent predictor of this condition. However, 25(OH)D level was independently associated with liver fibrosis even when adjusted for age, sex, body mass index, hemoglobin A1c, insulin, and homeostatic model assessment of insulin resistance (OR = 0.964 [0.935-0.993], P = 0.015). When patients were separated into subgroups by sex, a correlation between 25(OH)D and fibrosis was identified in the male group (OR = 0.969 [0.940-0.998], P = 0.038). Conclusions In conclusion, this multi-center, cross-sectional study in patients with T2DM showed that serum 25-hydroxy vitamin D level was strongly associated with liver fibrosis and this relationship was more pronounced in male patients. Clinical trial registration https://clinicaltrials.gov/ct2/show/, identifier NCT05597709.
Collapse
Affiliation(s)
- Nan Huang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Xianghui Su
- Department of Endocrinology, Changji Branch, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Ting Yu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaodong Wu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Bing Lu
- Department of Endocrinology, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Liqin Yao
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Maoyun Wang
- Department of Endocrinology, Baoying Hospital of Traditional Chinese Medicine, Yangzhou, China
| | - Yao Wang
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Wenxuan Wu
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Yingzhao Liu
- Department of Endocrinology, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Ting Yang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ruidong Gao
- Department of Endocrinology, Baoying Hospital of Traditional Chinese Medicine, Yangzhou, China
| | - Congqing Miao
- Department of Endocrinology, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| |
Collapse
|
28
|
Yu C, Ding C, Yu C, Bao H, Cheng X. Decoding the fatty liver-hyperuricemia link in the obese and nonobese hypertensive patients: insights from a cohort study. Sci Rep 2024; 14:29525. [PMID: 39604465 PMCID: PMC11603371 DOI: 10.1038/s41598-024-80895-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic-dysfunction-associated fatty liver disease (MAFLD) and serum uric acid are closely related to cardiovascular and cerebrovascular diseases. However, the causal association between MAFLD and serum uric acid remains unclear. A total of 3417 patients without hyperuricemia were included in the final analysis. MAFLD was defined as fatty liver index (FLI) ≥ 30. Multivariate Cox regression analysis was used to explore the association between FLI and new-onset hyperuricemia. Restricted cubic splines and threshold saturation effect analysis were used to detect nonlinear associations. The mean age was 62.8 ± 8.3 year, and 68.5% were women. A total of 738 (21.6%) hypertensive patients developed new-onset hyperuricemia, 388 (11.4%) new-onset hyperuricemia10 and 190 (5.6%) new-onset hyperuricemia20 during the 4-year midday follow-up period. In the fully adjusted model, compared with the Q1 (FLI ≤ 8.5) group, the risk of hyperuricemia increased by 56% (HR: 1.56; 95% CI: 1.02, 2.38) in the Q4 (FLI > 39.4) group, new-onset hyperuricemia10 increased by 108% (HR: 2.08; 95% CI: 1.15, 3.78), and new-onset hyperuricemia20 increased by 156% (HR: 2.56; 95% CI: 1.11, 5.94), respectively. Saturation effects showed a nonlinear association between FLI and new-onset hyperuricemia (p for log likelihood ratio test < 0.05). Subgroup analysis and stratified analysis showed that there had a significantly higher risk of new-onset hyperuricemia in the patients with normal body mass index (< 24 kg/m2) (p for interaction: 0.018) and non-central obesity (p for interaction: 0.024). MAFLD is an independent risk factor for hyperuricemia in hypertensive patients, especially in patients with normal body mass index and non-central obesity.
Collapse
Affiliation(s)
- Chuanli Yu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| | - Congcong Ding
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| | - Chao Yu
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| | - Huihui Bao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China.
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China.
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China.
| | - Xiaoshu Cheng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
- Center for Prevention and Treatment of Cardiovascular Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Cardiovascular Disease Clinical Medical Research Center, Nanchang, Jiangxi, China
- Jiangxi Sub-center of National Clinical Research Center for Cardiovascular Diseases, Nanchang, Jiangxi, China
| |
Collapse
|
29
|
Yuan C, Zhang C, Geng X, Feng C, Su Y, Wu Y, Wang Y, Chen L, Ding Q, Voortman T, Wang H, Zong G. Associations of an overall healthy lifestyle with the risk of metabolic dysfunction-associated fatty liver disease. BMC Public Health 2024; 24:3264. [PMID: 39587552 PMCID: PMC11587751 DOI: 10.1186/s12889-024-20663-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: 03/06/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) affects up to one-third of the global population. Since no approved pharmacotherapy for MAFLD is available, lifestyle modification remains the cornerstone of clinical care. Our study aims to evaluate the association of an overall healthy lifestyle with MAFLD risk. METHODS We conducted an analysis of 327,387 participants from UK biobank. An overall healthy lifestyle score including six evidence-based lifestyles (diet, alcohol consumption, physical activity, sedentary behavior, sleep, and smoking) was assessed by questionnaires. MAFLD and its subtypes were diagnosed by blood biochemistry, ICD codes, and medication information from touchscreen and verbal interview. The prevalence ratios (PRs) and risk ratios (RRs) were estimated by Poisson regression models with robust variance. RESULTS In the cross-sectional analysis, the PR (95% CI) was 0.83 (0.83 to 0.84) for MAFLD, and 0.83 (0.83 to 0.84) for MAFLD-overweight/obesity (MAFLD-O), 0.68 (0.66 to 0.70) for MAFLD-lean/normal weight and metabolic dysfunction (P-value for heterogeneity < 0.001), and 0.71 (0.71 to 0.72) for MAFLD-type 2 diabetes mellitus (MAFLD-T2D); and 0.68 (0.66 to 0.71) for dual (or more) etiology fatty liver disease (MAFLD-dual) and 0.83 (0.83 to 0.84) for single etiology MAFLD (MAFLD-single) (P-value for heterogeneity < 0.001) for one additional point in the overall healthy lifestyle score. During a median follow-up of 4.4 years, the RR (95% CI) was 0.83 (0.81 to 0.85) for MAFLD, and 0.83 (0.81 to 0.85) for MAFLD-O, 0.71 (0.62 to 0.81) for MAFLD-L, and 0.68 (0.64 to 0.72) for MAFLD-T2D (P-value for heterogeneity < 0.001); and 0.83 (0.81 to 0.85) for MAFLD-dual and 0.70 (0.58 to 0.85) for MAFLD-single (P-value for heterogeneity = 0.08) for one additional point in the overall healthy lifestyle score. These findings were validated in a prospective analysis among 15,721 participants with revisit data, and also supported by fatty liver index and proton density fat fraction data. CONCLUSIONS An overall healthy lifestyle that includes six evidence-based factors was strongly associated with lower MAFLD risk, especially the subtypes with multiple etiologies.
Collapse
Affiliation(s)
- Caimei Yuan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, PR China
| | - Chengjing Zhang
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Xin Geng
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
- National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China
- Ministry of Education (MOE) Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Naval Medical University, Shanghai, 200438, China
| | - Chengwu Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, PR China
| | - Yang Su
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, PR China
| | - Yinfan Wu
- Department of clinical nutrition, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200081, China
| | - Ying Wang
- Department of clinical nutrition, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200081, China
| | - Li Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, PR China
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, PR China
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, 3000 CA, The Netherlands
| | - Hongyang Wang
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
- National Center for Liver Cancer, Naval Medical University, Shanghai, 200438, China.
- Ministry of Education (MOE) Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Naval Medical University, Shanghai, 200438, China.
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, 225 Changhai Road, Shanghai, 200438, PR China.
| | - Geng Zong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, PR China.
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
30
|
Huang M, Zhang X, Zhou R, Song Y, Zhang J, Wu J. Advances in the study of oral microbiota and metabolism associated fatty liver disease: a systematic review. Front Cell Infect Microbiol 2024; 14:1491696. [PMID: 39600870 PMCID: PMC11588716 DOI: 10.3389/fcimb.2024.1491696] [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: 09/05/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Objective The oral microbiota is the second largest microbiota in the human body and has a significant impact on human health. Recent evidence suggests that dysbiosis of the oral microbiota may be associated with the development of metabolism-associated fatty liver disease (MAFLD). This review aimed to validate the relationship between oral microbial diversity and the development of MAFLD. Methods A systematic evaluation was performed based on PRISMA guidelines. Three independent reviewers searched for relevant literature in several databases, including PubMed/Medline, Web of Science, and Scopus, with a search date ranging from the establishment of the databases to June 2024. Results A total of 1278 publications were initially screened, including five cross-sectional studies, seven case-control studies, one cohort study, and one retrospective study. These studies included a total of 3335 patients with MAFLD, 254 patients with MASH, and 105 patients with liver cirrhosis. All 14 included studies concluded that there was a correlation or potential correlation between oral microbiota and MAFLD. Seven studies found that the composition of the oral microbiota in MAFLD patients differed from that of healthy controls, and specific oral bacteria may be associated with an increased incidence of MAFLD. At the phylum level, several studies found differences in the abundance of the phyla Firmicutes, Proteobacteria, and Clostridia compared to healthy controls. Additionally, a study on oral fungi found significant differences in the phyla Proteobacteria and in the genus Staphylococcus between patients with MAFLD and healthy controls. At the genus level, Porphyromonas was studied most frequently, with all 8 studies identifying infection with Porphyromonas as a significant risk factor for pathological progression in MAFLD. Furthermore, a dysbiosis in the ratio of Porphyromonas gingivalis./Porphyromonas anomalies may be an important marker of MAFLD progression. Conclusion There is an important association between the diversity of oral microbiota composition and MAFLD. This finding suggests the importance of oral health assessment and monitoring for the prevention or intervention of MAFLD.
Collapse
Affiliation(s)
- Mingming Huang
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Xinbi Zhang
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Rui Zhou
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Yingzhe Song
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Jing Zhang
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jian Wu
- School of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China
| |
Collapse
|
31
|
Ramírez-Mejía MM, Qi X, Abenavoli L, Méndez-Sánchez N. The myth of the stigma of fatty liver: What does the evidence show? Ann Hepatol 2024; 29:101535. [PMID: 39147131 DOI: 10.1016/j.aohep.2024.101535] [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: 04/16/2024] [Revised: 06/19/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Recent efforts to reclassify non-alcoholic fatty liver disease (NAFLD) as metabolic dysfunction-associated fatty liver disease (MAFLD) and metabolic dysfunction-associated steatotic liver disease (MASLD) are intended to divert attention to the metabolic basis of the disease rather than to alcohol consumption. This reclassification recognizes the role of obesity, sedentary lifestyles and poor dietary habits in the development of the disease, leading to a better understanding of its etiology. Nevertheless, the transition has posed its own challenges, particularly with regard to communication between patient and healthcare professional. Many healthcare professionals report difficulty in explaining the nuanced concepts, especially the term "steatosis". In addition, the change in terminology has not yet removed the stigma, with ongoing debates about the appropriateness of the terms "fatty" and "steatotic". Surveys suggest that while "obesity" may be perceived as more stigmatizing, the medical term "steatotic liver disease" is not considered as stigmatizing, indicating a disconnect in perceptions between healthcare professionals and patients.
Collapse
Affiliation(s)
- Mariana M Ramírez-Mejía
- Plan of Combined Studies in Medicine (PECEM-MD/PhD), Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico; Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Liaoning Province, PR China
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia of Catanzaro, Italy
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico; Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
32
|
Huang JF, Chang TJ, Yeh ML, Shen FC, Tai CM, Chen JF, Huang YH, Hsu CY, Cheng PN, Lin CL, Hung CH, Chen CC, Lee MH, Lee CC, Lin CW, Liu SC, Yang HI, Chien RN, Kuo CS, Peng CY, Chang ML, Huang CF, Yang YS, Yang HC, Lin HC, Ou HY, Liu CJ, Tseng CH, Kao JH, Chuang WL, Huang CN, Chen PJ, Wang CY, Yu ML. Clinical care guidance in patients with diabetes and metabolic dysfunction-associated steatotic liver disease: A joint consensus. Hepatol Commun 2024; 8:e0571. [PMID: 39470335 PMCID: PMC11524742 DOI: 10.1097/hc9.0000000000000571] [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: 06/22/2024] [Accepted: 08/05/2024] [Indexed: 10/30/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide, affecting >30% of the global population. Metabolic dysregulation, particularly insulin resistance and its subsequent manifestation as type 2 diabetes mellitus, serves as the fundamental pathogenesis of metabolic liver disease. Clinical evidence of the recent nomenclature evolution is accumulating. The interaction and impacts are bidirectional between MASLD and diabetes in terms of disease course, risk, and prognosis. Therefore, there is an urgent need to highlight the multifaceted links between MASLD and diabetes for both hepatologists and diabetologists. The surveillance strategy, risk stratification of management, and current therapeutic achievements of metabolic liver disease remain the major pillars in a clinical care setting. Therefore, the Taiwan Association for the Study of the Liver (TASL), Taiwanese Association of Diabetes Educators, and Diabetes Association of the Republic of China (Taiwan) collaboratively completed the first guidance in patients with diabetes and MASLD, which provides practical recommendations for patient care.
Collapse
Affiliation(s)
- Jee-Fu Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tien-Jyun Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Lun Yeh
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Feng-Chih Shen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Ming Tai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Jung-Fu Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Hsiang Huang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University Faculty of Medicine, Taipei, Taiwan
- Healthcare and Services Center and Therapeutic and Research Center of Liver Cancer, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Yao Hsu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taipei City Hospital Renai Branch, Taipei, Taiwan
| | - Pin-Nan Cheng
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Ling Lin
- Department of Internal Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Chao-Hung Hung
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Hepatogastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ching-Chu Chen
- Division of Endocrinology and Metabolism, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Mei-Hsuan Lee
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University Faculty of Medicine, Taipei, Taiwan
| | - Chun-Chuan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, Taipei, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, School of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Sung-Chen Liu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, Taipei, Taiwan
| | - Hwai-I Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Rong-Nan Chien
- Department of Gastroenterology and Hepatobiliary Disease, Linkou Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chin-Sung Kuo
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Yuan Peng
- Department of Internal Medicine, Center for Digestive Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Ling Chang
- Department of Gastroenterology and Hepatobiliary Disease, Linkou Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chung-Feng Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Sun Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Hung-Chih Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department and Graduate Institute of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Horng-Yih Ou
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Jen Liu
- Division of Gastroenterology & Hepatology, Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Chin-Hsiao Tseng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jia-Horng Kao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wan-Long Chuang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Ning Huang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung, Taiwan
| | - Pei-Jer Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Yuan Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine, Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
33
|
Zhang J, Hu J, Li Y, Zhou X, Ke Y, Chen Y. Serum Autotaxin Level Positively Associates with Metabolic-Associated Fatty Liver Disease and Hyperuricemia in Postmenopausal Women. Dig Dis 2024; 43:54-62. [PMID: 39442506 DOI: 10.1159/000542061] [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: 06/29/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Autotaxin (ATX) is an adipokine known to affect energy metabolism and lipid homeostasis. We aimed to evaluate serum ATX levels in metabolic-associated fatty liver disease (MAFLD) and other metabolic disorders in postmenopausal women. METHODS Postmenopausal women who received an annual health examination were included. The metabolic and demographic characteristics of the subjects were collected, including age, gender, weight, height, blood pressure, and biochemical parameters. Serum ATX level was determined by ELISA. RESULTS This cross-sectional includes 20 postmenopausal women and 20 age-paired healthy controls. MAFLD patients showed significant metabolic disturbance presented with increased body mass index (BMI), blood pressure (p < 0.001) and decreased high-density lipoprotein cholesterol (p < 0.05), as well as liver injury companied by elevated ALT (p < 0.05). Serum ATX levels were statistically higher in MAFLD (253.1 ± 52.1 vs. 202.2 ± 53.2 ng/mL; p < 0.01) and positively correlated with ALT (p < 0.001), γ-glutamyltransferase and BMI (p < 0.01), SBP and TG (p < 0.05). Higher ATX group demonstrated worsen metabolic states with greater proportion of MAFLD, higher BMI (p < 0.01), and ALT (p < 0.05). Logistic regression analysis revealed that serum ATX levels would positively independently predicted MAFLD (OR 1.049, 95% CI: 1.001-1.098, p < 0.05) with AUC of 0.763. Serum level of ATX is significantly elevated in hyperuricemia group (257.3 ± 60.9 vs. 214.5 ± 49.4 ng/mL; p < 0.05) and positively correlated with uric acid level (p < 0.01). Serum ATX would also act as diagnosing parameter of hyperuricemia with AUC of 0.706. CONCLUSIONS Among postmenopausal women, serum ATX level is significantly elevated in MAFLD and related to multiple metabolic characteristics, especially hyperuricemia, which would thus serve as a potential noninvasive biomarker as well as a therapeutic target.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiahui Hu
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yu Li
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xuefeng Zhou
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yini Ke
- Department of Rheumatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Chen
- Department of Gastroenterology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Souza M, Diaz I, Al-Sharif L. Liver and cardiovascular outcomes in lean non-alcoholic fatty liver disease: an updated systematic review and meta-analysis of about 1 million individuals. Hepatol Int 2024; 18:1396-1415. [PMID: 39117942 DOI: 10.1007/s12072-024-10716-z] [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: 04/20/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver disease (NAFLD) is present in lean people. However, the magnitude of the prognostic hepatic and cardiovascular risk in these patients compared to non-lean counterparts remains unclear. We aimed to investigate this topic, and to explore whether these risks change based on factors related to NAFLD severity. METHODS PubMed and Embase databases were searched for cohort studies (published through April 2024) that evaluated liver and cardiovascular (CV) outcomes in lean and non-lean individuals with NAFLD and reported unadjusted or adjusted data. We pooled risk ratios (RRs) or hazard ratios (HRs) using a random-effects modeling and performed subgroup and meta-regressions analyses. RESULTS We identified 22 studies with over 1 million NAFLD patients (13.0% were lean). Lean NAFLD showed a similar risk of liver-related events in unadjusted analysis (RR 1.08, 95% CI 0.79-1.49, I2 = 31%), but a higher risk in adjusted analysis (HR 1.66, 95% CI 1.17-2.36, I2 = 83%) compared to non-lean NAFLD. Lean NAFLD had a higher risk of liver-related mortality (RR 2.22, 95% CI 1.57-3.15, I2 = 0%; HR 2.26, 95% CI 1.14-4.51, I2 = 0%). For CV outcomes, lean NAFLD had a lower risk of any cardiovascular disease in unadjusted analysis (RR = 0.82, 95% CI 0.70-0.95, I2 = 88%), but similar risk in adjusted analysis (HR 0.89, 95% CI 0.77-1.02, I2 = 78%), and similar risk of cardiovascular mortality (RR 1.09, 95% CI 0.71-1.66, I2 = 85%; HR 1.26, 95% CI 0.89-1.78, I2 = 46%) compared to non-lean NAFLD. CONCLUSIONS Lean NAFLD patients have worse liver outcomes, but similar CV outcomes compared to non-lean NAFLD patients, highlighting the importance of monitoring both groups closely.
Collapse
Affiliation(s)
- Matheus Souza
- Department of Internal Medicine, Federal University of Rio de Janeiro, 255 Professor Rodolpho Paulo Rocco Av, Rio de Janeiro, 21941-913, Brazil.
| | - Ivanna Diaz
- Department of Internal Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | | |
Collapse
|
35
|
Cheng YM, Wang SW, Wang CC, Kao JH. Clinical characteristics of lean metabolic-associated fatty liver disease and the impact of concurrent diabetes mellitus. Tzu Chi Med J 2024; 36:425-432. [PMID: 39421499 PMCID: PMC11483085 DOI: 10.4103/tcmj.tcmj_253_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/20/2023] [Accepted: 01/17/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives Metabolic-associated fatty liver disease (MAFLD) was proposed in 2020 to replace the original term nonalcoholic fatty liver disease (NAFLD) with new diagnostic criteria. The disease risks of lean and overweight/obese MAFLD patients remain controversial. Materials and Methods The participants from the Taiwan biobank cohort were included. Advanced liver fibrosis is defined as NAFLD fibrosis score (NFS) >0.675. We use carotid plaques of duplex ultrasounds to diagnose atherosclerosis. Results A total of 20,058 participants (age 55.67 ± 10.32; males 37.6%) were included in the final analysis. Seven thousand eight hundred and forty-three (39.1%) participants were diagnosed with MAFLD. Of them, 965 (12.3%) were lean MAFLD patients. Among lean MAFLD patients, 25.6% were comorbid with diabetes mellitus (DM). Lean MAFLD patients were older and had higher percentages of females and DM than overweight/obese MAFLD patients. After propensity score matching for age and sex, they had lower levels of NFS but a higher percentage of carotid plaques. Among four subtypes of MAFLD including "lean with DM," "lean without DM," "overweight/obese with DM," and "overweight/obese without DM," logistic regression showed that "lean with DM" subjects had the highest risk of atherosclerosis and "overweight/obese with DM" subjects had the highest risk of advanced liver fibrosis in MAFLD patients. Conclusion The population-based study revealed that lean MAFLD patients make up 12.3% of all MAFLD patients, and they have a higher proportion of coexisting diabetes. Among lean MAFLD patients concurrent with diabetes, they have the highest risk of atherosclerosis and should receive special attention clinically.
Collapse
Affiliation(s)
- Yu-Ming Cheng
- Department of Gastroenterology and Hepatology, Tung’s Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shao-Wen Wang
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Chia-Chi Wang
- Department of Gastroenterology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Jia-Horng Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
36
|
Méndez-Sánchez N, Brouwer WP, Lammert F, Yilmaz Y. Metabolic dysfunction associated fatty liver disease in healthy weight individuals. Hepatol Int 2024; 18:884-896. [PMID: 39052203 PMCID: PMC11449956 DOI: 10.1007/s12072-024-10662-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/13/2024] [Indexed: 07/27/2024]
Abstract
Metabolic dysfunction associated fatty liver disease (MAFLD) is an increasing public health problem, affecting one third of the global population. Contrary to conventional wisdom, MAFLD is not exclusive to obese or overweight individuals. Epidemiological studies have revealed a remarkable prevalence among healthy weight individuals, leading investigations into the genetic, lifestyle, and dietary factors that contribute to the development of MAFLD in this population. This shift in perspective requires reconsideration of preventive strategies, diagnostic criteria and therapeutic approaches tailored to address the unique characteristics of MAFLD healthy weight individuals. It also underscores the importance of widespread awareness and education, within the medical community and among the general population, to promote a more inclusive understanding of liver metabolic disorders. With this review, we aim to provide a comprehensive exploration of MAFLD in healthy weight individuals, encompassing epidemiological, pathophysiological, and clinical aspects.
Collapse
Affiliation(s)
- Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic and Foundation, Mexico City, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Willem Pieter Brouwer
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Erasmus MC Transplant Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Frank Lammert
- Health Sciences, Hannover Medical School, Hannover, Germany
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| |
Collapse
|
37
|
Alarabi M, Pan Z, Romero-Gómez M, George J, Eslam M. Telomere length and mortality in lean MAFLD: the other face of metabolic adaptation. Hepatol Int 2024; 18:1448-1458. [PMID: 38900410 DOI: 10.1007/s12072-024-10701-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS Healthy weight (lean) patients with metabolic dysfunction-associated fatty liver disease (MAFLD) have a more favorable metabolic and histological profile in cross-sectional studies compared with their non-lean counterparts. Paradoxically, they also have higher overall mortality. The underpinning pathophysiology of this paradox is not understood. Telomere attrition is associated with increased mortality in various diseases. METHODS We investigated the role of telomere length in the pathogenesis of lean MAFLD in cohorts with biopsy-proven MAFLD (n = 303). We measured serum malondialdehyde (MDA) levels and hepatic 8-hydroxydeoxyguanosine (8-OHdG) and 4-hydroxy-2-nonenal (4-HNE) expression (reactive oxygen species (ROS) markers), growth/differentiation factor-15 (GDF-15) and tested the effect of H2O2 on telomere length and activity in hepatocyte cell lines. The association between leukocyte telomere length and mortality was examined. RESULTS Telomere length was significantly lower in patients with lean MAFLD (p < 0.001). They also demonstrated an increase in ROS levels and decreases in GDF-15. H2O2 induced telomere shortening and reducing telomere activity in hepatocyte cell lines. We subsequently confirmed that telomere length shortening at baseline is associated with increased hazards of all-cause mortality; the deleterious effect was more profound in lean people. CONCLUSION Differences in telomere length in part explain the increased mortality of lean compared to non-lean patients with MAFLD. The effect is in part mediated through ROS activation and provide opportunities for therapy.
Collapse
Affiliation(s)
- Mohammad Alarabi
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Sydney, 2145 NSW, Australia
| | - Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Sydney, 2145 NSW, Australia
| | - Manuel Romero-Gómez
- Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Sydney, 2145 NSW, Australia
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Sydney, 2145 NSW, Australia.
| |
Collapse
|
38
|
Quehenberger O, Armando AM, Cedeno TH, Loomba R, Sanyal AJ, Dennis EA. Novel eicosanoid signature in plasma provides diagnostic for metabolic dysfunction-associated steatotic liver disease. J Lipid Res 2024; 65:100647. [PMID: 39303979 PMCID: PMC11526069 DOI: 10.1016/j.jlr.2024.100647] [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: 04/23/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
There is a clinical need for a simple test implementable at the primary point of care to identify individuals with metabolic dysfunction-associated steatotic liver disease (MASLD) in the population. Blood plasma samples from adult patients with varying phenotypes of MASLD were used to identify a minimal set of lipid analytes reflective of underlying histologically confirmed MASLD. Samples were obtained from the NIDDK Nonalcoholic Steatohepatitis Clinical Research Network (NASH CRN) NAFLD Database prospective cohort study (MASLD group; N = 301). Samples of control subjects were obtained from cohort studies at the University of California San Diego (control group; N = 48). Plasma samples were utilized for targeted quantitation of circulating eicosanoids, related bioactive metabolites, and polyunsaturated fatty acids by ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS) lipidomics analysis. Bioinformatic approaches were used to discover a panel of bioactive lipids that can be used as a diagnostic tool to identify MASLD. The final panel of fifteen lipid metabolites consists of 12 eicosanoid metabolites and 3 free fatty acids that were identified to be predictive for MASLD by multivariate area under the receiver operating characteristics curve (AUROC) analysis. The panel was highly predictive for MASLD with an AUROC of 0.999 (95% CI = 0.986-1.0) with only one control misclassified. A validation study confirmed the resulting MASLD LIPIDOMICS SCORE, which may require a larger-scale prospective study to optimize. This predictive model should guide the development of a non-invasive "point-of-care" test to identify MASLD patients requiring further evaluation for the presence of metabolic dysfunction-associated steatohepatitis.
Collapse
Affiliation(s)
- Oswald Quehenberger
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| | - Aaron M Armando
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Tiffany H Cedeno
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Edward A Dennis
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA; Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
39
|
Zhou XD, Lonardo A, Pan CQ, Shapiro MD, Zheng MH. Clinical features and long-term outcomes of patients diagnosed with MASLD, MAFLD, or both. J Hepatol 2024; 81:e157-e159. [PMID: 38554846 DOI: 10.1016/j.jhep.2024.03.039] [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: 03/06/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Affiliation(s)
- Xiao-Dong Zhou
- Department of Cardiovascular Medicine, The Heart Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | | | - Calvin Q Pan
- Division of Gastroenterology and Hepatology, Department of Medicine, NYU Langone Health, New York University Grossman School of Medicine, New York, USA
| | - Michael D Shapiro
- Center for Prevention of Cardiovascular Disease, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Institute of Hepatology, Wenzhou Medical University, Wenzhou, 325000, China; Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, 325000, China.
| |
Collapse
|
40
|
Bae JH. Reply to correspondence on "Differences in liver and mortality outcomes of non-alcoholic fatty liver disease by race and ethnicity: A longitudinal real-world study". Clin Mol Hepatol 2024; 30:1047-1049. [PMID: 39159944 PMCID: PMC11540394 DOI: 10.3350/cmh.2024.0666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024] Open
Affiliation(s)
- Jae Hyun Bae
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
41
|
Eslam M, George J. MAFLD: from a disease framework to patient care. Hepatol Int 2024; 18:823-826. [PMID: 38886280 PMCID: PMC11449962 DOI: 10.1007/s12072-024-10685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/16/2024] [Indexed: 06/20/2024]
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia.
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
42
|
Zhang M, Chang D, Guan Q, Dong R, Zhang R, Zhang W, Wang H, Wang J. High-density lipoprotein cholesterol trajectory and new-onset metabolic dysfunction-associated fatty liver disease incidence: a longitudinal study. Diabetol Metab Syndr 2024; 16:223. [PMID: 39261925 PMCID: PMC11389356 DOI: 10.1186/s13098-024-01457-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 08/25/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Although high-density lipoprotein cholesterol (HDL-C) exerts a significant influence on the development of metabolic dysfunction-associated fatty liver disease (MAFLD), the association of dynamic changes in HDL-C levels with the risk of MAFLD remains unclear. Thus, the aim of the current study was to explore the association between the changing trajectories of HDL-C and new-onset MAFLD. The findings of this study may provide a theoretical basis for future personalized intervention and prevention targeting MAFLD. METHODS A total of 1507 participants who met the inclusion criteria were recruited from a community-based physical examination population in Nanjing, China from 2017 to 2021. Group-based trajectory models were constructed to determine the heterogeneous HDL-C trajectories. The incidence of MAFLD in each group in 2022 was followed up, and the Cox proportional hazards regression model was applied to investigate the associations between different HDL-C trajectories and the risk of new-onset MAFLD. RESULTS The incidences of MAFLD in the low-stable, moderate-stable, moderate-high-stable, and high-stable groups of HDL-C trajectory were 26.5%, 13.8%, 7.2% and 2.6%, respectively. The incidence rate of MAFLD in the order of the above trajectory groups exhibited a decreasing trend (χ2 = 72.55, Ptrend<0.001). After adjusting for confounders, the risk of MAFLD onset in HDL-C low-stable group was still 5.421 times (95%CI: 1.303-22.554, P = 0.020) higher than that in the high-stable group. Subgroup analyses of the combined (moderate high-stable and high-stable groups combined), moderate-stable and low-stable groups showed that sex, age, and overweight/obesity did not affect the association between HDL-C trajectory and MAFLD risk. CONCLUSIONS Persistently low HDL-C level is a risk factor for the onset of MAFLD. Long-term monitoring of HDL-C levels and timely intervention for those experiencing persistent declines are crucial for early prevention of MAFLD.
Collapse
Grants
- 2019, WSN-049 the Six Talent Peaks Project in Jiangsu Province, China
- 2019, WSN-049 the Six Talent Peaks Project in Jiangsu Province, China
- 2019, WSN-049 the Six Talent Peaks Project in Jiangsu Province, China
- 2019, WSN-049 the Six Talent Peaks Project in Jiangsu Province, China
- 2019, WSN-049 the Six Talent Peaks Project in Jiangsu Province, China
- Nursing Science, 2018, No.87 Priority Academic Program Development of Jiangsu Higher Education Institutions
- Nursing Science, 2018, No.87 Priority Academic Program Development of Jiangsu Higher Education Institutions
- Nursing Science, 2018, No.87 Priority Academic Program Development of Jiangsu Higher Education Institutions
- Nursing Science, 2018, No.87 Priority Academic Program Development of Jiangsu Higher Education Institutions
- Nursing Science, 2018, No.87 Priority Academic Program Development of Jiangsu Higher Education Institutions
Collapse
Affiliation(s)
- Mengting Zhang
- Department of Gastroenterology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Dongchun Chang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Qing Guan
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Rui Dong
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ru Zhang
- School of Nursing and Midwifery, Jiangsu College of Nursing, Huai'an, 223003, Jiangsu, China
| | - Wei Zhang
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, 200032, China
| | - Hongliang Wang
- Department of General Practice, Ninghai Road Community Health Service Center, Gulou District, Nanjing, 210024, Jiangsu, China
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
43
|
Halamy Pereira L, Barros FD, Andrade TGD, Oliveira Neto AAD, Nogueira CAV, Valezi AC. METABOLIC DYSFUNCTION-ASSOCIATED STEATOTIC LIVER DISEASE - ASSESSMENT OF PATIENTS WITH OBESITY AND METABOLIC SYNDROME - GUIDELINE FROM THE BRAZILIAN SOCIETY OF BARIATRIC AND METABOLIC SURGERY. ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2024; 37:e1821. [PMID: 39230102 PMCID: PMC11368249 DOI: 10.1590/0102-6720202400028e1821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease in the world and was recently renamed to emphasize its metabolic component. AIMS This article seeks to fill the gap in specific guidelines for patients with obesity and MASLD who will undergo bariatric surgery. METHODS A systematic search for guidelines was carried out on PubMed and Embase platforms. RESULTS A total of 544 articles were found, of which 11 were selected according to inclusion and exclusion criteria. All 11 guidelines are from clinical societies; therefore, they do not include some necessary interpretations for bariatric patients. CONCLUSIONS We recommend that every patient undergoing bariatric and metabolic surgery be screened initially with the Fibrosis-4 (FIB-4) score, followed by transient hepatic elastography (vibration-controlled transient elastography, VCTE), especially for those with FIB-4>1.3. However, interpreting VCTE results in obese patients requires further studies to define the actual cutoff values. Enhanced Liver Fibrosis® shows promise but its availability is limited. The indication for liver biopsy during surgery needs to be individualized but it is recommended for those with changes in FIB-4 and/or VCTE. Family screening is recommended for relatives of young patients with already advanced fibrosis. Liver transplantation is an option for patients with advanced MASLD but the optimal timing for bariatric surgery with transplantation is still unclear. Regular follow-up and VCTE examination are recommended to monitor disease progression after surgery.
Collapse
Affiliation(s)
- Leonardo Halamy Pereira
- Universidade Federal Fluminense, Department of Specialized and General Surgery - Niterói (RJ), Brazil
| | - Fernando de Barros
- Universidade Federal Fluminense, Department of Specialized and General Surgery - Niterói (RJ), Brazil
| | | | | | | | | |
Collapse
|
44
|
Procyk G, Jaworski J, Gąsecka A, Filipiak KJ, Borovac JA. Metabolic dysfunction-associated steatotic liver disease - A new indication for sodium-glucose Co-transporter-2 inhibitors. Adv Med Sci 2024; 69:407-415. [PMID: 39260740 DOI: 10.1016/j.advms.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/09/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has been proposed as a new name for the previous non-alcoholic fatty liver disease (NAFLD). There are some differences between MASLD and NAFLD, e.g., diagnostic criteria. MASLD is a hepatic steatosis without harmful alcohol consumption and is caused by metabolic factors. The prevalence of MASLD varies amongst different populations. The change in lifestyle plays a fundamental role in MASLD management, while there is no registered pharmacotherapy in this indication. Sodium-glucose co-transporter 2 inhibitors (SGLT2i) have been suggested to have a beneficial effect on hepatic steatosis, hence, they have been widely investigated as potential therapeutics in MASLD. In this review, we aimed to thoroughly summarize current evidence from original research about the effects of SGLT2i use on MASLD. Almost all discussed studies advocate using SGLT2i in MASLD because of their beneficial effects. It includes the loss of body weight, which is beneficial per se, and the improvement in hepatic parameters. Most importantly, steatosis reduction has been observed in patients using SGLT2i. We highly recommend further research in this field, which we believe will eventually lead to a new indication for SGLT2i, i.e., MASLD.
Collapse
Affiliation(s)
- Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland; Doctoral School, Medical University of Warsaw, Warsaw, Poland.
| | - Jakub Jaworski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof J Filipiak
- Institute of Clinical Science, Maria Sklodowska-Curie Medical Academy, Warsaw, Poland; Department of Hypertension, Angiology and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Josip A Borovac
- Cardiovascular Diseases Department, University Hospital of Split, Split, Croatia
| |
Collapse
|
45
|
Pan Z, Derbala M, AlNaamani K, Ghazinian H, Fan JG, Eslam M. MAFLD criteria are better than MASLD criteria at predicting the risk of chronic kidney disease. Ann Hepatol 2024; 29:101512. [PMID: 38710473 DOI: 10.1016/j.aohep.2024.101512] [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: 04/17/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION AND OBJECTIVES Fatty liver disease is a multisystem disease. Metabolic dysfunction-associated fatty liver disease (MAFLD) is a more accurate indicator of chronic kidney disease (CKD) than nonalcoholic fatty liver disease (NAFLD). However, the relationship between recently defined metabolic dysfunction-associated steatotic liver disease (MASLD) and CKD is currently unclear. The objective of this cross-sectional study was to investigate the prevalence of CKD and albuminuria among individuals diagnosed with either MAFLD or MASLD. PATIENTS AND METHODS This study involved 5,492 participants who provided biochemical marker and liver ultrasound data from the U.S. National Health and Nutrition Examination Survey (2017-2020). Multiple logistic regression analyses were conducted to assess the independent associations of nonoverlapping MAFLD and MASLD with the presence of CKD or albuminuria (urinary albumin-to-creatinine ratio ≥ 3 mg/mmol). RESULTS MAFLD and MASLD were identified in 47% and 44.5% of the participants, respectively. Individuals with MAFLD-only had a greater prevalence of CKD (24.7% vs. 8.3 %, P < 0.006) and albuminuria (18.6% vs. 5%, P < 0.01) than did those with MASLD-only. Importantly, after adjusting for factors such as sex, age, ethnicity, and alcohol use, it was demonstrated that individuals in the MAFLD-only group had a 4.73-fold greater likelihood of having prevalent CKD than those in the MASLD-only group (P < 0.03). CONCLUSIONS The MAFLD criteria better identify patients with CKD than do the MASLD criteria. Therefore, it is suggested that the MASLD criteria be reconsidered, as currently, the justification for changing from MAFLD to MASLD criteria may not be appropriate.
Collapse
Affiliation(s)
- Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| | - Moutaz Derbala
- Gastroenterology and Hepatology Department, Hamad Medical Corporation, Doha, Qatar
| | - Khalid AlNaamani
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Armed Forces Hospital, Muscat, Oman
| | - Hasmik Ghazinian
- Gastroenterology and Hepatology Department, Medical Scientific Center, Yerevan, Armenia
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia.
| |
Collapse
|
46
|
Chen J, Lu L, Nie X, Li J, Chen T, Li S. Associations of exposure to perchlorate, thiocyanate, and nitrate with metabolic dysfunction–associated steatotic liver disease: Evidence from a population-based cross-sectional study in the United States. JOURNAL OF CLEANER PRODUCTION 2024; 469:143156. [DOI: 10.1016/j.jclepro.2024.143156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
47
|
Jun JH, Du K, Dutta RK, Maeso-Diaz R, Oh SH, Wang L, Gao G, Ferreira A, Hill J, Pullen SS, Diehl AM. The senescence-associated secretome of Hedgehog-deficient hepatocytes drives MASLD progression. J Clin Invest 2024; 134:e180310. [PMID: 39190624 PMCID: PMC11444248 DOI: 10.1172/jci180310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
The burden of senescent hepatocytes correlates with the severity of metabolic dysfunction-associated steatotic liver disease (MASLD), but the mechanisms driving senescence and how it exacerbates MASLD are poorly understood. Hepatocytes experience lipotoxicity and become senescent when Smoothened (Smo) is deleted to disrupt Hedgehog signaling. We aimed to determine whether the secretomes of Smo-deficient hepatocytes perpetuate senescence to drive MASLD progression. RNA-Seq analysis of liver samples from human and murine cohorts with MASLD confirmed that hepatocyte populations in MASLD livers were depleted of Smo+ cells and enriched with senescent cells. When fed a choline-deficient, amino acid-restricted high-fat diet (CDA-HFD) to induce MASLD, Smo- mice had lower antioxidant markers and developed worse DNA damage, senescence, steatohepatitis, and fibrosis than did Smo+ mice. Sera and hepatocyte-conditioned medium from Smo- mice were depleted of thymidine phosphorylase (TP), a protein that maintains mitochondrial fitness. Treating Smo- hepatocytes with TP reduced senescence and lipotoxicity, whereas inhibiting TP in Smo+ hepatocytes had the opposite effect and exacerbated hepatocyte senescence, steatohepatitis, and fibrosis in CDA-HFD-fed mice. We conclude that inhibition of Hedgehog signaling in hepatocytes promoted MASLD by suppressing hepatocyte production of proteins that prevent lipotoxicity and senescence.
Collapse
Affiliation(s)
- Ji Hye Jun
- Division of Gastroenterology, Department of Medicine and
| | - Kuo Du
- Division of Gastroenterology, Department of Medicine and
| | | | | | - Seh Hoon Oh
- Division of Gastroenterology, Department of Medicine and
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Guannan Gao
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Ana Ferreira
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Jon Hill
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Steven S Pullen
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, USA
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine and
| |
Collapse
|
48
|
Luo Q, Liu G, Li Q, Lu J, Zheng W, Huang Y, Li C. Novel insights into causal effects of maternal nonalcoholic fatty liver disease on adverse pregnancy outcomes: evidence from Human Genetics and Mendelian Randomization Study. Eur J Clin Nutr 2024:10.1038/s41430-024-01489-7. [PMID: 39147848 DOI: 10.1038/s41430-024-01489-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Observational studies have associated nonalcoholic fatty liver disease (NAFLD) with adverse pregnancy events, but findings show heterogeneity, leaving the causal direction and mediating pathways unclear. We aimed to investigate the causal relation between NAFLD and various pregnancy events, and to elucidate the underlying mediating pathways while determining the proportion of this correlation that is mediated through these pathways. METHODS A genome-wide association study involving over 6 million participants employing Mendelian randomization (MR) and mediation analysis was performed. The study used genetically predicted NAFLD as exposures and cardiometabolic traits as mediators, with various adverse pregnancy events as outcomes. The main analysis was performed using the inverse variance weighted (IVW) approach, while sensitivity analyses included the weighted median, weighted mode, MR-Egger, and MR-PRESSO methods. Mediation analyses were performed using a two-step MR framework. RESULTS In this MR cohort study, NAFLD was found to be strongly associated with elevated risks of GDM (P = 0.019 for the discovery dataset, P < 0.001 for the discovery dataset) and HDPs, including any HDP (P < 0.001 for the both datasets), gestational hypertension (P = 0.007 for the discovery dataset, P < 0.001 for the discovery dataset), and pre-eclampsia or eclampsia (P = 0.040 for the discovery dataset, P < 0.001 for the discovery dataset). However, no significant associations were found with hemorrhage in early pregnancy, postpartum hemorrhage, preterm birth, or offspring birthweight for both datasets. Cardiometabolic traits played a significant mediating role in these associations, rather than solely acting as confounding factors. CONCLUSIONS This study provided evidence supporting a correlation between NAFLD and a higher risk of adverse pregnancy events and introduces some new insights. These findings may inform preventions and interventions for remediating adverse pregnancy outcomes attributable to NAFLD.
Collapse
Affiliation(s)
- Qiuyan Luo
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Guoting Liu
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Qiulan Li
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Jinghong Lu
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Wenjing Zheng
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China
| | - Yukui Huang
- Department of Obstetrics and Gynecology, Nanning Maternity and Child Health Hospital, Nanning, PR China.
| | - Cun Li
- Department of Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, PR China.
| |
Collapse
|
49
|
Zhai D, Xu S, Liu H, Tong X. Nonalcoholic or metabolic-associated fatty liver disease and colorectal polyps: evidence from meta-analysis and two-sample Mendelian randomization. Front Genet 2024; 15:1422827. [PMID: 39184353 PMCID: PMC11341362 DOI: 10.3389/fgene.2024.1422827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Nonalcoholic or metabolism-associated fatty liver disease (NAFLD or MAFLD) and colorectal polyps are chronic conditions strongly linked to lifestyle factors. However, the precise causal link between NAFLD or MAFLD and the development of colorectal polyps is not yet fully understood. This study aimed to evaluate the association between NAFLD or MAFLD and the risk of colorectal polyps based on a meta-analysis and two-sample Mendelian randomization (MR) analyses. Methods PubMed, Embase, Cochrane Library databases were searched for eligible studies to be included in the meta-analysis. We conducted a thorough search of the PubMed, Embase, and Cochrane Library databases to identify eligible studies prior to 22 March 2024. Subgroup analyses were performed based on sex, age, and geographical region. Causality between NAFLD/MAFLD and colorectal polyps was explored by using two-sample Mendelian randomization (MR) analyses. Results Based on an analysis of 17 studies encompassed within this meta-analysis, a significant correlation was identified between the presence of NAFLD/MAFLD and elevated incidence of colorectal polyps (NAFLD: OR = 1.57, 95% CI: 1.43-1.73, I2 = 38%, p = 0.06; MAFLD: OR = 1.67, 95% CI: 1.40-2.00, I2 = 77%, p = 0.002). However, current evidence does not support a causal relationship between NAFLD/MAFLD and the prevalence of colorectal polyps (OR = 0.9998315, 95% CI: 0.9987566-1.000907, P = 0.7587638). Conclusion NAFLD/MAFLD demonstrated a significant positive correlation with an elevated risk of developing colorectal polyps. However, the MR analysis suggested that no causal relationship existed between NAFLD/MAFLD and colorectal polyps. Therefore, further research is required to identify the underlying mechanism of causal link between these diseases.
Collapse
Affiliation(s)
- Dong Zhai
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, China
| | - Sumei Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Haoge Liu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Xiaojuan Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
50
|
Wang K, Margolis S, Cho JM, Wang S, Arianpour B, Jabalera A, Yin J, Hong W, Zhang Y, Zhao P, Zhu E, Reddy S, Hsiai TK. Non-Invasive Detection of Early-Stage Fatty Liver Disease via an On-Skin Impedance Sensor and Attention-Based Deep Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400596. [PMID: 38887178 PMCID: PMC11336938 DOI: 10.1002/advs.202400596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/17/2024] [Indexed: 06/20/2024]
Abstract
Early-stage nonalcoholic fatty liver disease (NAFLD) is a silent condition, with most cases going undiagnosed, potentially progressing to liver cirrhosis and cancer. A non-invasive and cost-effective detection method for early-stage NAFLD detection is a public health priority but challenging. In this study, an adhesive, soft on-skin sensor with low electrode-skin contact impedance for early-stage NAFLD detection is fabricated. A method is developed to synthesize platinum nanoparticles and reduced graphene quantum dots onto the on-skin sensor to reduce electrode-skin contact impedance by increasing double-layer capacitance, thereby enhancing detection accuracy. Furthermore, an attention-based deep learning algorithm is introduced to differentiate impedance signals associated with early-stage NAFLD in high-fat-diet-fed low-density lipoprotein receptor knockout (Ldlr-/-) mice compared to healthy controls. The integration of an adhesive, soft on-skin sensor with low electrode-skin contact impedance and the attention-based deep learning algorithm significantly enhances the detection accuracy for early-stage NAFLD, achieving a rate above 97.5% with an area under the receiver operating characteristic curve (AUC) of 1.0. The findings present a non-invasive approach for early-stage NAFLD detection and display a strategy for improved early detection through on-skin electronics and deep learning.
Collapse
Affiliation(s)
- Kaidong Wang
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
- Department of MedicineGreater Los Angeles Veterans Affairs (VA) Healthcare SystemLos AngelesCA90073USA
| | - Samuel Margolis
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jae Min Cho
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Shaolei Wang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Brian Arianpour
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Alejandro Jabalera
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Junyi Yin
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Wen Hong
- Department of Materials Science and EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Yaran Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Peng Zhao
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Enbo Zhu
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Materials Science and EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Srinivasa Reddy
- Department of Molecular and Medical PharmacologyUniversity of California Los AngelesLos AngelesCA90095USA
| | - Tzung K. Hsiai
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
- Department of MedicineGreater Los Angeles Veterans Affairs (VA) Healthcare SystemLos AngelesCA90073USA
| |
Collapse
|