1
|
Kang J, Zhu JQ, Wang Y, He Q. Effect of Immunosuppressive Regimens on Metabolic Dysfunction-associated Fatty Liver Disease Following Liver Transplantation. J Clin Exp Hepatol 2025; 15:102387. [PMID: 39268481 PMCID: PMC11388780 DOI: 10.1016/j.jceh.2024.102387] [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: 04/17/2024] [Accepted: 07/31/2024] [Indexed: 09/15/2024] Open
Abstract
Background Metabolic dysfunction-associated fatty liver disease has been linked to negative outcomes in patients with end-stage liver disease following liver transplantation. However, the influence of immunosuppressive regimens on it has not been explored. Methods A retrospective analysis was conducted using the preoperative and postoperative data from patients with end-stage liver disease. The study compared three different groups: tacrolimus-based group, sirolimus-based group, and combined tacrolimus- and sirolimus-based regimens. Binary logistic regression analysis was employed to identify risk factors for metabolic dysfunction-associated fatty liver disease. Results A total of 171 patients participated in the study, consisting of 127 males and 44 females, with a mean age of 49.6 years. The prevalence of posttransplant metabolic dysfunction-associated fatty liver disease was 29.23%. Among the three groups, there were 111 liver transplant recipients in the tacrolimus-based group, 28 in the sirolimus-based group, and 32 in the combination group. A statistically significant difference was observed in the incidence of metabolic dysfunction-associated fatty liver disease (P < 0.05), whereas the other preoperative and postoperative parameters showed no significant differences. Multivariate analysis revealed that a low-calorie diet (95% confidence intervals: 0.15-0.90, P = 0.021) and a combination of tacrolimus- and sirolimus-based immunosuppressive regimen (95% confidence intervals: 1.01-2.77, P = 0.046) were associated with lower risk of posttransplant metabolic dysfunction-associated fatty liver disease. Conclusions Our study indicates that implementing a low-calorie diet and utilizing a combination of tacrolimus- and sirolimus-based immunosuppressive regimen can effectively lower the risk of posttransplant metabolic dysfunction-associated fatty liver disease following liver transplantation.
Collapse
Affiliation(s)
- Jing Kang
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Ji-Qiao Zhu
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Yan Wang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Qiang He
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Organ Transplant Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| |
Collapse
|
2
|
Miranda J, Key Wakate Teruya A, Leão Filho H, Lahan-Martins D, Tamura Sttefano Guimarães C, de Paula Reis Guimarães V, Ide Yamauchi F, Blasbalg R, Velloni FG. Diffuse and focal liver fat: advanced imaging techniques and diagnostic insights. Abdom Radiol (NY) 2024; 49:4437-4462. [PMID: 38896247 DOI: 10.1007/s00261-024-04407-4] [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: 04/16/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
The fatty liver disease represents a complex, multifaceted challenge, requiring a multidisciplinary approach for effective management and research. This article uses conventional and advanced imaging techniques to explore the etiology, imaging patterns, and quantification methods of hepatic steatosis. Particular emphasis is placed on the challenges and advancements in the imaging diagnostics of fatty liver disease. Techniques such as ultrasound, CT, MRI, and elastography are indispensable for providing deep insights into the liver's fat content. These modalities not only distinguish between diffuse and focal steatosis but also help identify accompanying conditions, such as inflammation and fibrosis, which are critical for accurate diagnosis and management.
Collapse
Affiliation(s)
- Joao Miranda
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
- Department of Radiology, University of São Paulo, R. Dr. Ovídio Pires de Campos, 75-Cerqueira César, São Paulo, SP, 05403-010, Brazil.
| | - Alexandre Key Wakate Teruya
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Hilton Leão Filho
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Daniel Lahan-Martins
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
- Departament of Radiology-FCM, State University of Campinas (UNICAMP), R. Tessália Vieira de Camargo, 126 Cidade Universitária, Campinas, SP, 13083-887, Brazil
| | - Cássia Tamura Sttefano Guimarães
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Vivianne de Paula Reis Guimarães
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Fernando Ide Yamauchi
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Roberto Blasbalg
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Fernanda Garozzo Velloni
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| |
Collapse
|
3
|
Zhang C, Cao L, Xu B, Zhang W. Interaction between trouble sleeping and diabetes on metabolic dysfunction-associated fatty liver disease and liver fibrosis in adults results from the National Health and Nutrition Examination Survey 2017-2018. Eur J Gastroenterol Hepatol 2024; 36:1437-1446. [PMID: 39373628 PMCID: PMC11527372 DOI: 10.1097/meg.0000000000002860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD), trouble sleeping, and diabetes, as major public health problems, were closely related. The study examined the interaction between trouble sleeping and diabetes on MAFLD and liver fibrosis in adults with MAFLD. METHODS The data were obtained from the National Health and Nutrition Examination Survey 2017-2018. Multivariate logistic regression model and subgroup analyses were conducted to assess the relationship between either trouble sleeping or diabetes on MAFLD and liver fibrosis. Relative excess risk due to interaction (RERI), attributable proportion of interaction (AP), and synergy index (S) were utilized to assess the additive interaction. RESULTS Ultimately, 3747 participants were included, with 2229 known MAFLD subjects. Compared with participants without diabetes, those with diabetes had a higher risk of MAFLD [odds ratio (OR) = 5.55; 95% confidence interval (CI) = 4.07-7.56] and liver fibrosis risk (OR = 3.61; 95% CI = 2.67-4.89). We also found a significant association of trouble sleeping with an increased risk of MAFLD (OR = 1.54; 95% CI = 1.17-2.02) and liver fibrosis risk (OR = 1.51; 95% CI = 1.06-2.16), compared with those without trouble sleeping. Moreover, there was a significant interaction between diabetes and trouble sleeping on MAFLD [RERI = 1.76 (95% CI: -0.22 to 3.73), AP = 0.35 (95% CI: 0.08-0.63), S = 1.80 (95% CI: 1.02-3.16)] and liver fibrosis risk [RERI = 1.79 (95% CI: 0.37-3.21), AP = 0.44 (95% CI: 0.20-0.69), S = 2.44 (95% CI: 1.18-5.08)]. CONCLUSION The findings highlight that trouble sleeping and diabetes had a synergistic effect on MAFLD and liver cirrhosis. The study highlights the importance of addressing both trouble sleeping and diabetes management in adults to mitigate the risk of MAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Cui Zhang
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| | - Lili Cao
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| | - Bo Xu
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| | - Wei Zhang
- Department of Oncology, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
4
|
Guney-Coskun M, Basaranoglu M. Interplay of gut microbiota, glucagon-like peptide receptor agonists, and nutrition: New frontiers in metabolic dysfunction-associated steatotic liver disease therapy. World J Gastroenterol 2024; 30:4682-4688. [DOI: 10.3748/wjg.v30.i43.4682] [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: 08/19/2024] [Revised: 09/20/2024] [Accepted: 10/14/2024] [Indexed: 10/31/2024] Open
Abstract
The gut-liver axis plays a crucial role in the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Key metabolites, including lipopolysaccharides, short-chain fatty acids (SCFAs), bile acids, and beneficial gut bacteria such as Bifidobacterium and Lactobacillus, are pivotal in this process. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) show promise in managing MASLD by promoting weight loss, enhancing insulin secretion, and improving liver health. They restore gut-liver axis functionality, and their effects are amplified through dietary modifications and gut microbiome-targeted therapies. Emerging research highlights the interplay between GLP-1 RAs and gut microbiota, indicating that the gut microbiome significantly influences therapeutic outcomes. Metabolites produced by gut bacteria, can stimulate glucagon-like peptide-1 (GLP-1) secretion, further improving metabolic health. Integrating dietary interventions with GLP-1 RA treatment may enhance liver health by modulating the gut microbiota-SCFAs-GLP-1 pathway. Future research is needed to understand personalized effects, with prebiotics and probiotics offering treatment avenues for MASLD.
Collapse
Affiliation(s)
- Merve Guney-Coskun
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Medipol University, Istanbul 34810, Türkiye
- Department of Nutrition and Dietetics, Graduate School of Health Sciences, Istanbul Medipol University, Istanbul 34810, Türkiye
| | - Metin Basaranoglu
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Bezmialem Vakif University, Istanbul 34093, Türkiye
| |
Collapse
|
5
|
Yang YL, Chuang YT, Huang YH. MicroRNA 29a alleviates mitochondrial stress in diet-induced NAFLD by inhibiting the MAVS pathway. Eur J Pharmacol 2024; 982:176955. [PMID: 39209098 DOI: 10.1016/j.ejphar.2024.176955] [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: 05/14/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder characterized by fat accumulation in the liver. This leads to aggravated hepatocyte inflammation due to impaired mitochondrial function, mitochondrial double-stranded RNA (mt-dsRNA) release, elevated oxidative stress, and reactive oxygen species (ROS) production. MicroRNA-29a (miR-29a) is used to reduce hepatic fibrosis in cases of cholestatic liver damage and lessen the severity of non-alcoholic steatohepatitis in animal studies by influencing mitochondrial protein balance. However, the effectiveness of miR-29a in diminishing mt-dsRNA-induced exacerbation of NAFLD remains poorly understood, particularly in the context of a Western diet (WD). Our results have found that mice with increased miR-29a levels and fed a WD showed notably decreased serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol, and low-density lipoprotein cholesterol levels. They also experienced less weight gain and lower final body and liver weights. In addition, overexpression of miR-29a reduced the severity of fibrosis, alleviated hepatic oxidative stress, misfolded protein aggregates, and the release of mt-dsRNA. Moreover, miR-29a attenuated the innate immune mitochondrial antiviral-signaling protein (MAVS) pathway response. In vitro, the research using HepG2 cells confirmed that miR-29a reduces MAVS expression and decreases the release of mt-dsRNA and superoxide initiated by palmitic acid (PA). Analysis of luciferase activity further established that the specific binding of miR-29a to the 3'UTR of MAVS led to a repression of its expression. In conclusion, these groundbreaking findings underscore the potential of miR-29a in improving the treatment of NAFLD and liver steatofibrosis by inhibiting the MAVS signaling pathway.
Collapse
Affiliation(s)
- Ya-Ling Yang
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 833, Taiwan
| | - Yuan-Ting Chuang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital Chang, and Chang Gung University College of Medicine, Kaohsiung, 833, Taiwan
| | - Ying-Hsien Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital Chang, and Chang Gung University College of Medicine, Kaohsiung, 833, Taiwan.
| |
Collapse
|
6
|
Pontikoglou CG, Filippatos TD, Matheakakis A, Papadaki HA. Steatotic liver disease in the context of hematological malignancies and anti-neoplastic chemotherapy. Metabolism 2024; 160:156000. [PMID: 39142602 DOI: 10.1016/j.metabol.2024.156000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The rising prevalence of obesity-related illnesses, such as metabolic steatotic liver disease (MASLD), represents a significant global public health concern. This disease affects approximately 30 % of the adult population and is the result of metabolic abnormalities rather than alcohol consumption. Additionally, MASLD is associated with an increased risk of cardiovascular disease (CVD), chronic liver disease, and a variety of cancers, particularly gastrointestinal cancers. Clonal hematopoiesis (CH) is a biological state characterized by the expansion of a population of blood cells derived from a single mutated hematopoietic stem cell. The presence of CH in the absence of a diagnosed blood disorder or cytopenia is known as clonal hematopoiesis of indeterminate potential (CHIP), which itself increases the risk of hematological malignancies and CVD. Steatotic liver disease may also complicate the clinical course of cancer patients receiving antineoplastic agents, a condition referred to as chemotherapy induced steatohepatitis (CASH). This review will present an outline of the various aspects of MASLD, including complications. Furthermore, it will summarize the existing knowledge on the emerging association between CHIP and MASLD and present the available data on patient cases with concurrent MASLD and hematological neoplasms. Finally, it will provide a brief overview of the chemotherapeutic drugs associated with CASH, the underlying pathophysiologic mechanisms and their clinical implications.
Collapse
Affiliation(s)
- Charalampos G Pontikoglou
- Department of Hematology, University Hospital of Heraklion, & School of Medicine of the University of Crete, Crete, Greece
| | - Theodosios D Filippatos
- Department of Internal Medicine, University Hospital of Heraklion, & School of Medicine of the University of Crete, Crete, Greece
| | - Angelos Matheakakis
- Department of Hematology, University Hospital of Heraklion, & School of Medicine of the University of Crete, Crete, Greece
| | - Helen A Papadaki
- Department of Hematology, University Hospital of Heraklion, & School of Medicine of the University of Crete, Crete, Greece.
| |
Collapse
|
7
|
Bhardwaj M, Mazumder PM. The gut-liver axis: emerging mechanisms and therapeutic approaches for nonalcoholic fatty liver disease and type 2 diabetes mellitus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8421-8443. [PMID: 38861011 DOI: 10.1007/s00210-024-03204-6] [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: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), more appropriately known as metabolic (dysfunction) associated fatty liver disease (MAFLD), a prevalent condition in type 2 diabetes mellitus (T2DM) patients, is a complex condition involving hepatic lipid accumulation, inflammation, and liver fibrosis. The gut-liver axis is closely linked to metabolic dysfunction, insulin resistance, inflammation, and oxidative stress that are leading to the cooccurrence of MAFLD and T2DM cardiovascular diseases (CVDs). The purpose of this review is to raise awareness about the role of the gut-liver axis in the progression of MAFLD, T2DM and CVDs with a critical analysis of available treatment options for T2DM and MAFLD and their impact on cardiovascular health. This study analysed over 100 articles on this topic, using online searches and predefined keywords, to understand and summarise published research. Numerous studies have shown a strong correlation between gut dysfunction, particularly the gut microbiota and its metabolites, and the occurrence and progression of MAFLD and type 2 diabetes mellitus (T2DM). Herein, this article also examines the impact of the gut-liver axis on MAFLD, T2DM, and related complications, focusing on the role of gut microbiota dysbiosis in insulin resistance, T2DM and obesity-related cardiovascular complications. The study suggests potential treatment targets for MAFLD linked to T2DM, focusing on cardiovascular outcomes and the molecular mechanism of the gut-liver axis, as gut microbiota dysbiosis contributes to obesity-related metabolic abnormalities.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India.
| |
Collapse
|
8
|
Tan W, Zhang J, Chen L, Wang Y, Chen R, Zhang H, Liang F. Copper homeostasis and cuproptosis-related genes: Therapeutic perspectives in non-alcoholic fatty liver disease. Diabetes Obes Metab 2024; 26:4830-4845. [PMID: 39233500 DOI: 10.1111/dom.15846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/06/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), a metabolic-associated fatty liver disease, has become the most common chronic liver disease worldwide. Recently, the discovery of cuproptosis, a newly identified mode of cell death, further highlighted the importance of copper in maintaining metabolic homeostasis. An increasing number of studies have confirmed that liver copper metabolism is closely related to the pathogenesis of NAFLD. However, the relationship between NAFLD and copper metabolism, especially cuproptosis, remains unclear. In this review, we aim to summarize the current understanding of copper metabolism and its dysregulation, particularly the role of copper metabolism dysregulation in the pathogenesis of NAFLD. More importantly, this review emphasizes potential gene-targeted therapeutic strategies, challenges and the future of cuproptosis-related genes in the treatment of NAFLD. This review aims to provide innovative therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Wangjing Tan
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Junli Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Chen
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Yayuan Wang
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Rui Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiming Zhang
- Department of Oncology, Integrated Traditional Chinese and Western Medicine, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxia Liang
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Acupuncture and Moxibustion Department, Affiliated Hospital of Hubei University of Chinese Medicine(Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| |
Collapse
|
9
|
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
|
10
|
Xu H, Liu Y, Wei Y. Impact of metabolic dysfunction-associated fatty liver disease on the outcomes following laparoscopic hepatectomy for hepatocellular carcinoma. Surg Endosc 2024; 38:6456-6463. [PMID: 39266759 DOI: 10.1007/s00464-024-11239-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/29/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND The impact of metabolic dysfunction-associated fatty liver disease (MAFLD) on laparoscopic liver resection (LLR) for hepatocellular carcinoma (HCC) remains unclear. This study aimed to compare the outcomes of LLR for MAFLD-HCC and Non-MAFLD-HCC. METHODS Patients with HCC who received LLR between October 2017 and July 2021 were enrolled. Inverse probability of treatment weighting (IPTW) was used to generate adjusted comparisons. Both short- and long-term outcomes were evaluated accordingly. RESULTS A total of 887 patients were enrolled, with 140 in MAFLD group and 747 in Non-MAFLD group. After IPTW adjustment, baseline factors were well matched. The MAFLD group was associated with more blood loss (210 vs 150 ml, p = 0.022), but with similar postoperative hospital stays and complication rates. The 1- and 3-year overall survival rates were 97.4% and 92.5% in MAFLD group, and 97.5% and 88.3% in Non-MAFLD group, respectively (p = 0.14). The 1- and 3-year disease-free survival rates were 84.8% and 62.9% in MAFLD group, and 80.2% and 58.8% in Non-MAFLD group, respectively (p = 0.31). CONCLUSIONS LLR for MAFLD-HCC was associated with more blood loss but with comparable postoperative recovery and long-term survival compared with Non-MAFLD-HCC patients. LLR is feasible and safe for HCC patients with MAFLD background.
Collapse
Affiliation(s)
- Hongwei Xu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guo Xue Road, Chengdu, 610041, Sichuan Province, China
| | - Yani Liu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guo Xue Road, Chengdu, 610041, Sichuan Province, China
| | - Yonggang Wei
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, 37 Guo Xue Road, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
11
|
Kim EY, Lee YJ, Kwon YJ, Lee JW. Age at menopause and risk of metabolic dysfunction-associated fatty liver disease: A 14-year cohort study. Dig Liver Dis 2024; 56:1880-1886. [PMID: 38763798 DOI: 10.1016/j.dld.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUNDS & AIMS Menopause, characterized by a sudden decline in estrogen levels, has significant effects on women's health, especially when it occurs early. This study aimed to investigate the associations between menopausal age and incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) using a large cohort and a long-term follow-up. METHODS Menopausal age was categorized into four groups (G1-4 [<40, 40-44, 45-49, and ≥50 years, respectively]). Cox proportional hazards regression analysis was used to assess the risk of developing MAFLD during the follow-up period according to the menopausal age categories. RESULTS A total of 1,888 participants were included in the final analysis and followed for a median period of 12.3 years. The unadjusted hazard ratios (95 % CIs) for the incidence of new-onset MAFLD were 1.11 (0.93-1.32), 1.15 (0.90-1.47), and 1.52 (1.12-2.07) in G3, G2, and G1, respectively, compared with that in G4. After adjusting for confounders, the hazard ratio (95 % CIs) for the incidence of new-onset MAFLD was 1.40 (1.00-1.95) in G1 compared with that in G4. CONCLUSION The risk of developing MAFLD was higher in women with premature menopause (<40 years) than in those with menopause aged ≥50 years.
Collapse
Affiliation(s)
- Ehn-Young Kim
- Department of Family Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
| | - Yae-Ji Lee
- Department of Biostatistics and Computing, Yonsei University, Seoul 03722, Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, 363, Dongbaekjukjeon-daero, Giheung-gu, Yongin-si 16995, Gyeonggi-do, Korea.
| | - Ji-Won Lee
- Department of Family Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1, Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea; Institute for Innovation in Digital Healthcare, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
12
|
Xiao J, Zhang X, Chang L, Yu H, Sun L, Zhu C, He Q. Associations of four surrogate insulin resistance indexes with non-alcoholic steatohepatitis in Chinese patients with obesity: a cross-sectional study. Endocrine 2024; 86:546-555. [PMID: 38814373 DOI: 10.1007/s12020-024-03888-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024]
Abstract
OBJECTIVES This study was designed to evaluate the association of four surrogate indexes of IR with NASH in patients with obesity. METHODS A total of 270 patients who underwent bariatric surgery, were included in this cross-sectional study. NASH was diagnosed based on liver biopsies. Binary logistics regression analyses were performed to assess the associations of four surrogate indexes of IR (HOMA-IR, Matsuda index, TyG, and TG/HDL-C) with NASH in patients with obesity. The restricted cubic spline was used to assess the dose-response associations of surrogate indexes of IR with NASH after adjusting for confounding factors. RESULTS NASH was diagnosed in 136 patients, with a prevalence of 50.37%. Compared with tertile 1, the fully adjusted ORs (95% CIs) of NASH for tertile 3 were 2.711(1.113-6.608) and 0.297 (0.152-0.579) for TyG and Matsuda index. Consistently, per SD increment of TyG were still significantly associated with 64% increased risks of NASH, and per SD increment of Matsuda index were still significantly associated with 38% decreased risks of NASH. In contrast, no significant associations were found between HOMA-IR and TG/HDL-C and the risk of NASH in patients with obesity (all P > 0.05). After adjusting covariates in restricted cubic splines, the risk of NASH decreased with the increment of Matsuda Index levels (P-nonlinear = 0.442, P-overall = 0.007) and with the decrement of TyG levels (P-nonlinear = 0.004, P-overall = 0.001). CONCLUSIONS In patients with obesity, TyG and Matsuda index were independently related to the risk of NASH after adjustment for traditional risk factors. In addition, compared with HOMA-IR and TG/HDL-C, the Matsuda index and TyG may be more suitable for NASH prediction in patients with obesity.
Collapse
Affiliation(s)
- Jinfeng Xiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Endocrinology and Metabolism, Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Xinxin Zhang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Lina Chang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hong Yu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Longhao Sun
- Department of General surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Chonggui Zhu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
13
|
Hosseini Shabanan S, Martins VF, Wolfson T, Weeks JT, Ceriani L, Behling C, Chernyak V, El Kaffas A, Borhani AA, Han A, Wang K, Fowler KJ, Sirlin CB. MASLD: What We Have Learned and Where We Need to Go-A Call to Action. Radiographics 2024; 44:e240048. [PMID: 39418184 DOI: 10.1148/rg.240048] [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: 10/19/2024]
Abstract
Since its introduction in 1980, fatty liver disease (now termed metabolic dysfunction-associated steatotic liver disease [MASLD]) has grown in prevalence significantly, paralleling the rise of obesity worldwide. While MASLD has been the subject of extensive research leading to significant progress in the understanding of its pathophysiology and progression factors, several gaps in knowledge remain. In this pictorial review, the authors present the latest insights into MASLD, covering its recent nomenclature change, spectrum of disease, epidemiology, morbidity, and mortality. The authors also discuss current qualitative and quantitative imaging methods for assessing and monitoring MASLD. Last, they propose six unsolved challenges in MASLD assessment, which they term the proliferation, reproducibility, reporting, needle-in-the-haystack, availability, and knowledge problems. These challenges offer opportunities for the radiology community to proactively contribute to their resolution. The authors conclude with a call to action for the entire radiology community to claim a seat at the table, collaborate with other societies, and commit to advancing the development, validation, dissemination, and accessibility of the imaging technologies required to combat the looming health care crisis of MASLD.
Collapse
Affiliation(s)
- Sedighe Hosseini Shabanan
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Vitor F Martins
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Tanya Wolfson
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Jake T Weeks
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Lael Ceriani
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Cynthia Behling
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Victoria Chernyak
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Ahmed El Kaffas
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Amir A Borhani
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Aiguo Han
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Kang Wang
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Kathryn J Fowler
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| | - Claude B Sirlin
- From the Department of Radiology, UC San Diego Altman Clinical and Translational Research Institute Liver Imaging Group, University of California San Diego, 9452 Medical Center Dr, La Jolla, CA 92037 (S.H.S., V.F.M., T.W., J.T.W., L.C., K.J.F., C.B.S.); Pacific Rim Pathology, San Diego, Calif (C.B.); Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY (V.C.); Department of Radiology, Stanford University School of Medicine, Stanford, Calif (A.E.K.); Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Ill (A.A.B.); Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Va (A.H.); and Department of Radiology, University of California San Francisco, Calif (K.W.)
| |
Collapse
|
14
|
Zhang H, Dong X, Zhu L, Tang FS. Elafibranor: A promising treatment for alcoholic liver disease, metabolic-associated fatty liver disease, and cholestatic liver disease. World J Gastroenterol 2024; 30:4393-4398. [PMID: 39494094 PMCID: PMC11525860 DOI: 10.3748/wjg.v30.i40.4393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/16/2024] Open
Abstract
Liver diseases pose a significant threat to human health. Although effective therapeutic agents exist for some liver diseases, there remains a critical need for advancements in research to address the gaps in treatment options and improve patient outcomes. This article reviews the assessment of Elafibranor's effects on liver fibrosis and intestinal barrier function in a mouse model of alcoholic liver disease (ALD), as reported by Koizumi et al in the World Journal of Gastroenterology. We summarize the impact and mechanisms of Elafibranor on ALD, metabolic-associated fatty liver disease, and cholestatic liver disease based on current research. We also explore its potential as a dual agonist of PPARα/δ, which is undergoing Phase III clinical trials for metabolic-associated steatohepatitis. Our goal is to stimulate further investigation into Elafibranor's use for preventing and treating these liver diseases and to provide insights for its clinical application.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| | - Xuan Dong
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| | - Lei Zhu
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| | - Fu-Shan Tang
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
- Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi Medical University, Zunyi 563006, Guizhou Province, China
| |
Collapse
|
15
|
Qiao Y, Chen S, Liu Y, Li L, He L, Han Z, Liu F, Cheng Z, Yu X, Yu J, Pang C, Liang P. Ultrasound-guided percutaneous microwave ablation for metabolic-associated fatty liver disease-related hepatocellular carcinoma (HCC) versus hepatitis virus B-related HCC: a propensity score matching study. Int J Hyperthermia 2024; 41:2419912. [PMID: 39462518 DOI: 10.1080/02656736.2024.2419912] [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: 05/11/2024] [Revised: 07/29/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVES To compare the long-term outcomes of microwave ablation (MWA) for primary hepatocellular carcinoma (HCC) in patients with metabolic-associated fatty liver disease (MAFLD) with those infected by hepatitis virus B (HBV). METHODS The clinical data of HCC patients under the treatment of MWA were analyzed retrospectively between 2010 and 2021 at Chinese PLA General Hospital. Patients were divided into MAFLD-HCC and HBV-HCC group according to the chronic liver disease etiology. The propensity score matching (PSM) was performed to reduce the interference of confounders. The primary outcomes were overall survival (OS), recurrence-free survival (RFS), cancer-specific survival (CSS) and intrahepatic distant recurrence (IDR). RESULTS A total of 648 patients (age range, 18-91 years) with 1019 lesions were enrolled including 601 with HBV-HCC and 47 with MAFLD-HCC. After a variable ratio of 1:n ≤ 4 PSM, 100 patients were included in the HBV-HCC and 41 in the MAFLD-HCC group. No statistical differences in OS and CSS (p = 0.880 and p = 0.980, respectively) were observed between the two groups in the matched cohort, while MAFLD-HCC exhibited better RFS and lower IDR rates compared to HBV-HCC (p = 0.043 and p = 0.041, respectively). Additionally, MAFLD-HCC generated lower ascending range in the liver function indexes like ΔALT (46.7 vs. 98.5, p < 0.001), ΔTBIL (1.9 vs. 7.5, p = 0.001) and ΔAST (38.1 vs. 148.6, p < 0.001) than HBV-HCC after MWA. CONCLUSIONS MWA is an effective treatment for HCC patients with MAFLD. The recurrence prognosis of MAFLD-HCC was better than HBV-HCC and the degree of liver injury after MWA was lower.
Collapse
Affiliation(s)
- Yannan Qiao
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Sitong Chen
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Yang Liu
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lu Li
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Liting He
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zhiyu Han
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fangyi Liu
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhigang Cheng
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jie Yu
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chuan Pang
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ping Liang
- Department of Interventional Ultrasound, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Guo Z, Yao Z, Huang B, Wu D, Li Y, Chen X, Lu Y, Wang L, Lv W. MAFLD-related hepatocellular carcinoma: Exploring the potent combination of immunotherapy and molecular targeted therapy. Int Immunopharmacol 2024; 140:112821. [PMID: 39088919 DOI: 10.1016/j.intimp.2024.112821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common cause of cancer-related mortality and morbidity globally, and with the prevalence of metabolic-related diseases, the incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) related hepatocellular carcinoma (MAFLD-HCC) continues to rise with the limited efficacy of conventional treatments, which has created a major challenge for HCC surveillance. Immune checkpoint inhibitors (ICIs) and molecularly targeted drugs offer new hope for advanced MAFLD-HCC, but the evidence for the use of both types of therapy in this type of tumour is still insufficient. Theoretically, the combination of immunotherapy, which awakens the body's anti-tumour immunity, and targeted therapies, which directly block key molecular events driving malignant progression in HCC, is expected to produce synergistic effects. In this review, we will discuss the progress of immunotherapy and molecular targeted therapy in MAFLD-HCC and look forward to the opportunities and challenges of the combination therapy.
Collapse
Affiliation(s)
- Ziwei Guo
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People 's Hospital, Beijing 100044, China
| | - Bohao Huang
- Beijing University of Chinese Medicine, Beijing 100105, China
| | - Dongjie Wu
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yanbo Li
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaohan Chen
- Department of Hematology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanping Lu
- Department of Hepatology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518100, China.
| | - Li Wang
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Wenliang Lv
- Department of Infection, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
17
|
Chen M, Guo C, Ouyang K, Liu N. Diagnostic role of the fibrosis-4 index and nonalcoholic fatty liver disease fibrosis score as a noninvasive tool for liver fibrosis scoring. Medicine (Baltimore) 2024; 103:e40214. [PMID: 39470560 PMCID: PMC11521016 DOI: 10.1097/md.0000000000040214] [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: 06/27/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by liver fibrosis, which serves as a crucial indicator of its progression and prognosis. Owing to the limitations of biopsy, which is the gold standard for measuring liver fibrosis, a reliable and noninvasive marker is required. We evaluated the diagnostic role of the fibrosis-4 (FIB-4) index and nonalcoholic fatty liver disease fibrosis score (NFS) in patients with NAFLD with varying severities of liver fibrosis. The FIB-4 index and NFS were calculated using laboratory data from 121 patients who underwent liver biopsies between January 2022 and December 2023. The results were compared with those of the Scheuer scoring system for liver biopsies (F0, F1 + F2, and F3 + F4) to determine the sensitivity and specificity of the FIB-4 index and the liver disease fibrosis score in detecting and staging liver fibrosis. Twenty-one patients had advanced fibrosis (F3-F4), and 100 had minimal or mild fibrosis (F0-F2). The degree of liver fibrosis increased with decreased albumin, alanine aminotransferase and platelet count levels, and increasing age. Receiver operating characteristic curve analysis for the FIB-4 index and NFS revealed that the areas under the curve for the FIB-4 index and NFS were 0.895 (95% confidence interval: 0.836-0.954) and 0.882 (95% confidence interval: 0.813-0.952), respectively. The FIB-4 indices showed 95.24% sensitivity at a cutoff point of 1.30, and 85% specificity at a cutoff point of 2.67, while the NFS indices showed 95.24% sensitivity at -1.455 cutoff point and 95% specificity at a cutoff point of 0.676. The FIB-4 index and NFS may replace biopsy for the detection of fibrosis in patients with NAFLD.
Collapse
Affiliation(s)
- Mingxi Chen
- Department of Infectious Disease and Liver Disease, The Second Hospital of Nanjing, Nanjing, Jiangsu, China
| | - Chang Guo
- Department of Internal Medicine, Shandong Rehabilitation Hospital, Jinan, Shandong, China
| | - Ke Ouyang
- Department of Infectious Disease and Liver Disease, The Second Hospital of Nanjing, Nanjing, Jiangsu, China
| | - Na Liu
- Department of Infectious Disease and Liver Disease, The Second Hospital of Nanjing, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Yan J, Nie Y, Zhang S. Letter: MASLD in people with HIV exhibits higher fibrosis stage despite lower disease activity than in matched controls. Aliment Pharmacol Ther 2024. [PMID: 39449251 DOI: 10.1111/apt.18282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
LINKED CONTENTThis article is linked to Allende et al paper. To view this article, visit https://doi.org/10.1111/apt.18236
Collapse
Affiliation(s)
- Junbin Yan
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), Zhejiang Chinese Medical University, Hangzhou, China
| | - Yunmeng Nie
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (The Xin Hua Hospital of Zhejiang Province), Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
19
|
Torre E, Di Matteo S, Martinotti C, Bruno GM, Goglia U, Testino G, Rebora A, Bottaro LC, Colombo GL. Economic Impact of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in Italy. Analysis and Perspectives. CLINICOECONOMICS AND OUTCOMES RESEARCH 2024; 16:773-784. [PMID: 39469584 PMCID: PMC11514691 DOI: 10.2147/ceor.s472446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024] Open
Abstract
Background Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multisystem condition destined to become pandemic in the coming decades. This study aimed at evaluating the economic impact of MASLD in the Italian population from the Italian National Healthcare Service (NHS) perspective. Methods The economic impact of the MASLD was assessed by developing a calculation model in Microsoft Excel® from the Italian NHS perspective, considering healthcare resources and direct costs. The target population was based on the prevalence data. Through a literature search, complications of MASLD were identified, including MASH, with relative risk of evolution into CC, DCC, HCC, T2DM, cardiovascular diseases, in particular AMI and stroke, CKD, and CRC. The differential impact between complication development in the population with MASLD and the same sample size population without-MASLD was evaluated. Differential risk data, mortality rates, and event unit costs were drawn from the published international literature. Frequency and cost data were applied to the total target population, the total annual costs and mortality data, referring to the two arms, were then calculated, and the differential value was obtained. Results Based on an estimated 11,546,370 MASLD target population, an annual illness impact of €12,251,631,822 was calculated, corresponding to a difference of €7,731,674,054 compared with the same sample size without MASLD. Moreover, the MASLD population is expected to experience 13,438 additional deaths annually. Conclusion The growing epidemiological impact of MASLD and its complications represent a huge economic burden for healthcare services worldwide. An integrated approach, including changes in lifestyle behaviors, will be the first step. Specific drugs for MASLD are not yet available; however, studies are underway, and combined pharmaceutical therapies may be an inevitable choice to achieve adequate control of MASLD and its complications.
Collapse
Affiliation(s)
- Enrico Torre
- Diabetology and Metabolic Diseases Unit - ASL3, Genoa, Italy
| | - Sergio Di Matteo
- Center of Research, SAVE Studi - Health Economics and Outcomes Research, Milan, Italy
| | - Chiara Martinotti
- Center of Research, SAVE Studi - Health Economics and Outcomes Research, Milan, Italy
| | | | | | | | - Alberto Rebora
- Diabetology and Metabolic Diseases Unit - ASL3, Genoa, Italy
| | | | | |
Collapse
|
20
|
Younossi ZM, Stepanova M. Stigma Among Chinese Speaking Patients with Non-alcoholic Fatty Liver Disease and Their Providers. J Hepatol 2024:S0168-8278(24)02647-3. [PMID: 39454690 DOI: 10.1016/j.jhep.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Affiliation(s)
- Zobair M Younossi
- The Global Liver Council, Washington DC, USA; Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Disease, Washington DC, USA
| | - Maria Stepanova
- The Global Liver Council, Washington DC, USA; Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, VA, USA; Center for Outcomes Research in Liver Disease, Washington DC, USA
| |
Collapse
|
21
|
Dong F, He K, Zhang S, Song K, Jiang L, Hu LP, Li Q, Zhang XL, Zhang N, Li BT, Zhu LL, Li J, Feng M, Gao Y, Chen J, Hu X, Wang J, Jiang C, Wang C, Zhu HH, Da LT, Ji J, Zhang ZG, Bao Z, Jiang SH. SSRI antidepressant citalopram reverses the Warburg effect to inhibit hepatocellular carcinoma by directly targeting GLUT1. Cell Rep 2024; 43:114818. [PMID: 39388353 DOI: 10.1016/j.celrep.2024.114818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/20/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) have shown promise in cancer therapy, particularly for hepatocellular carcinoma (HCC), but their molecular targets and mechanisms remain unclear. Here, we show that SSRIs exhibit significant anti-HCC effects independent of their classical target, the serotonin reuptake transporter (SERT). Using global inverse gene expression profiling, drug affinity responsive target stability assays, and in silico molecular docking, we demonstrate that citalopram targets glucose transporter 1 (GLUT1), resulting in reduced glycolytic flux. A mutant GLUT1 variant at the citalopram binding site (E380) diminishes the drug's inhibitory effects on the Warburg effect and tumor growth. In preclinical models, citalopram dampens the growth of GLUT1high liver tumors and displays a synergistic effect with anti-PD-1 therapy. Retrospective analysis reveals that SSRI use correlates with a lower risk of metastasis among patients with HCC. Our study describes a role for SSRIs in cancer metabolism, establishing a rationale for their repurposing as potential anti-cancer drugs for HCC.
Collapse
Affiliation(s)
- Fangyuan Dong
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Kang He
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kaiyuan Song
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Luju Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qing Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xue-Li Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Naiqi Zhang
- Center for Primary Health Care Research, Lund University, Region Skåne, Sweden
| | - Bo-Tai Li
- Shanghai Immune Therapy Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Li-Li Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jun Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingxuan Feng
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yunchen Gao
- Shanghai United International School Qingpu Campus, Shanghai 201799, China
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Xiaona Hu
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Jiaofeng Wang
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Chongyi Jiang
- Department of General Surgery, Hepato-Biliary-Pancreatic Center, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Helen He Zhu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lin-Tai Da
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianguang Ji
- Center for Primary Health Care Research, Lund University, Region Skåne, Sweden; Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhijun Bao
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; Shanghai Institute of Geriatrics and Gerontology, Shanghai 200040, China; Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
22
|
Wu Y, Zhang Z, Cai H, Zhang W, Zhang L, Li Z, Yang L, Chen Y, Corner TP, Song Z, Yue J, Yang F, Li X, Schofield CJ, Zhang X. Discovery of ZG-2305, an Orally Bioavailable Factor Inhibiting HIF Inhibitor for the Treatment of Obesity and Fatty Liver Disease. J Med Chem 2024. [PMID: 39432709 DOI: 10.1021/acs.jmedchem.4c01698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Genetic loss of the 2-oxoglutarate oxygenase factor inhibiting hypoxia-inducible factor (FIH) enhances both glycolysis and aerobic metabolism. FIH is thus a potential target for adiposity control and improving hepatic steatosis. We describe development of a series of novel, potent, and selective FIH inhibitors that occupy both the FIH catalytic site and a recently defined tyrosine conformational-flip pocket. ZG-2305, with a Ki of 79.6 nM for FIH, manifests 38-fold selectivity over the hypoxia-inducible factor (HIF) prolyl hydroxylase PHD2. Oral administration of ZG-2305 in the western-diet induced obesity mouse model results in improved lipid accumulation and recovery from abnormal body weight/hepatic steatosis. Amelioration of nonalcoholic steatohepatitis (NASH) related pathological phenotypes in the HF-CDAA-diet induced NASH mouse model was observed. Preliminary preclinical studies indicate ZG-2305 has good pharmacokinetic properties and an acceptable safety profile. The results imply ZG-2305 is a promising candidate for treatment of obesity and fatty liver disease.
Collapse
Affiliation(s)
- Yue Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Zewei Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Haiping Cai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Weiqing Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Linjian Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Zhihong Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Le Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yafen Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Thomas P Corner
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Zhe Song
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Yue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Fulai Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Xiaojin Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
23
|
Li P, Wang T, Qiu H, Zhang R, Yu C, Wang J. 6-Gingerol Inhibits De Novo Lipogenesis by Targeting Stearoyl-CoA Desaturase to Alleviate Fructose-Induced Hepatic Steatosis. Int J Mol Sci 2024; 25:11289. [PMID: 39457074 PMCID: PMC11508832 DOI: 10.3390/ijms252011289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), also known as non-alcoholic fatty liver disease (NAFLD), is a worldwide liver disease without definitive or widely used therapeutic drugs in clinical practice. In this study, we confirm that 6-gingerol (6-G), an active ingredient of ginger (Zingiber officinale Roscoe) in traditional Chinese medicine (TCM), can alleviate fructose-induced hepatic steatosis. It was found that 6-G significantly decreased hyperlipidemia caused by high-fructose diets (HFD) in rats, and reversed the increase in hepatic de novo lipogenesis (DNL) and triglyceride (TG) levels induced by HFD, both in vivo and in vitro. Mechanistically, chemical proteomics and cellular thermal shift assay (CETSA)-proteomics approaches revealed that stearoyl-CoA desaturase (SCD) is a direct binding target of 6-G, which was confirmed by further CETSA assay and molecular docking. Meanwhile, it was found that 6-G could not alter SCD expression (in either mRNA or protein levels), but inhibited SCD activity (decreasing the desaturation levels of fatty acids) in HFD-fed rats. Furthermore, SCD deficiency mimicked the ability of 6-G to reduce lipid accumulation in HF-induced HepG2 cells, and impaired the improvement in hepatic steatosis brought about by 6-G treatment in HFD supplemented with oleic acid diet-induced SCD1 knockout mice. Taken together, our present study demonstrated that 6-G inhibits DNL by targeting SCD to alleviate fructose diet-induced hepatic steatosis.
Collapse
Affiliation(s)
- Pan Li
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Tingting Wang
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Hongmei Qiu
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Ruoyu Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China;
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Chao Yu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing Medical University, Chongqing 400016, China; (P.L.); (T.W.)
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China;
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing 400016, China;
| |
Collapse
|
24
|
Pang J, Yin L, Jiang W, Wang H, Cheng Q, Jiang Z, Cao Y, Zhu X, Li B, Qian S, Yin X, Wang T, Lu Q, Yang T. Sirt1-mediated deacetylation of PGC-1α alleviated hepatic steatosis in type 2 diabetes mellitus via improving mitochondrial fatty acid oxidation. Cell Signal 2024; 124:111478. [PMID: 39428026 DOI: 10.1016/j.cellsig.2024.111478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Being activated by deacetylation, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) has become an important regulator of metabolic-related diseases. The activation of Sirtuin 1 (Sirt1) by resveratrol was likely to deacetylate PGC-1α. However, the role of deacetylated PGC-1α in the alleviation of activated Sirt1 on type 2 diabetes mellitus (T2DM)-related fatty liver disease (FLD) remained unexplored. The aim of this study was to investigate the potential impact of Sirt1-mediated deacetylation of PGC-1α on T2DM-associated FLD and its underlying mechanisms. Our findings revealed that, along with the decreased Sirt1, the levels of acetylated PGC-1α were up-regulated in hepatocytes co-stimulated with high glucose (HG) and free fatty acids (FFA). Down-regulated Sirt1 inactivated PGC-1α by inhibiting its deacetylation, while activating Sirt1 improved hepatic injury by reducing lipid droplet accumulation through the deacetylation of PGC-1α. However, the beneficial effects of Sirt1 activation on hepatic steatosis were inhibited by PGC-1α antagonist in vitro. Mechanistically, activating Sirt1 enhanced mitochondrial function by promoting PGC-1α activity, thereby facilitating hepatic fatty acid oxidation (FAO). In conclusion, Sirt1-mediated deacetylation of PGC-1α mitigated hepatic lipotoxicity by enhancing mitochondrial FAO, which contributed to the restoration of mitochondrial function in T2DM. The activation of Sirt1-mediated PGC-1α deacetylation might represent a promising therapeutic approach for T2DM-associated FLD.
Collapse
Affiliation(s)
- Jiale Pang
- Department of Pharmacy, Jintan Affiliated Hospital of Jiangsu University, Changzhou 213200, China
| | - Longxiang Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenjie Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiyan Wang
- Department of Biochemistry, Graduate School of Inovative Life Science, University of Toyama, Toyama 930-0194, Japan
| | - Qian Cheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China, Pharmaceutical University, Nanjing 210009, China
| | - Yanjuan Cao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xia Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Baojing Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Sitong Qian
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Tao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China; Department of Pharmacy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China.
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
25
|
Belka M, Gostyńska-Stawna A, Stawny M, Krajka-Kuźniak V. Activation of Nrf2 and FXR via Natural Compounds in Liver Inflammatory Disease. Int J Mol Sci 2024; 25:11213. [PMID: 39456994 PMCID: PMC11508530 DOI: 10.3390/ijms252011213] [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: 09/27/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Liver inflammation is frequently linked to oxidative stress and dysregulation of bile acid and fatty acid metabolism. This review focuses on the farnesoid X receptor (FXR), a critical regulator of bile acid homeostasis, and its interaction with the nuclear factor erythroid 2-related factor 2 (Nrf2), a key modulator of cellular defense against oxidative stress. The review explores the interplay between FXR and Nrf2 in liver inflammatory diseases, highlighting the potential therapeutic effects of natural FXR agonists. Specifically, compounds such as auraptene, cafestol, curcumin, fargesone A, hesperidin, lycopene, oleanolic acid, resveratrol, rutin, ursolic acid, and withaferin A are reviewed for their ability to modulate both the FXR and Nrf2 pathways. This article discusses their potential to alleviate liver inflammation, oxidative stress, and damage in diseases such as metabolic-associated fatty liver disease (MAFLD), cholestatic liver injury, and viral hepatitis. In addition, we address the molecular mechanisms driving liver inflammation, including oxidative stress, immune responses, and bile acid accumulation, while also summarizing relevant experimental models. This review emphasizes the promising therapeutic potential of targeting both the Nrf2 and FXR pathways using natural compounds, paving the way for future treatments for liver diseases. Finally, the limitations of the clinical application were indicated, and further research directions were proposed.
Collapse
Affiliation(s)
- Marta Belka
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Aleksandra Gostyńska-Stawna
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| |
Collapse
|
26
|
Zhuang P, Ao Y, Liu X, Ye H, Li H, Wan X, Zhang Y, Jiao J. Circulating fatty acids and risk of severe non-alcoholic fatty liver disease in the UK biobank: a prospective cohort of 116 223 individuals. Food Funct 2024; 15:10527-10538. [PMID: 39370886 DOI: 10.1039/d4fo01182a] [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: 10/08/2024]
Abstract
Fatty acid (FA) metabolism plays an important role in the development of nonalcoholic fatty liver disease (NAFLD). However, data on the relationship between circulating FAs and NAFLD risk are limited. This study aims to assess the associations between specific circulating FAs and severe NAFLD risk among the general population. Overall 116 223 participants without NAFLD and other liver diseases from the UK Biobank were enrolled between 2006 and 2010 and were followed up until the end of 2021. Plasma concentrations of saturated fatty acids (SFAs), monounsaturated fatty acids (MUFAs), and polyunsaturated fatty acids (PUFAs) were analyzed using an NMR-based biomarker profiling platform. Hazard ratios (HRs) and 95% confidence intervals (CIs) of NAFLD risk were estimated using Cox proportional-hazard models adjusted for other potential confounders. During a mean follow-up of 12.3 years, we documented 1394 cases of severe NAFLD. After multivariate adjustment, plasma SFAs and MUFAs were associated with a higher risk of severe NAFLD, whereas plasma n-3 PUFAs, n-6 PUFAs, and linoleic acid (LA) were associated with a lower risk. As compared with the lowest quartile, HRs (95% CIs) of severe NAFLD risk in the highest quartiles were 1.85 (1.45-2.36) for SFAs, 1.74 (1.23-2.44) for MUFAs, 0.79 (0.65-0.97) for n-3 PUFAs, 0.68 (0.48-0.96) for n-6 PUFAs, and 0.73 (0.54-0.99) for LA. The significant relationships were mainly mediated by serum TG for SFAs, HDL-C for MUFAs and n-6 PUFAs, and C-reactive protein for n-3 PUFAs. Plasma SFAs were associated with a more pronounced increase in the risk of severe NAFLD among participants with fewer SFA-associated alleles (P interaction = 0.032). Dietary recommendations for reducing plasma SFAs and MUFAs while increasing n-3 and n-6 PUFAs may be protective for severe NAFLD, which could be mediated by lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Pan Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Yang Ao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Xiaohui Liu
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Hao Ye
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Haoyu Li
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xuzhi Wan
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| | - Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jingjing Jiao
- Department of Endocrinology, The Second Affiliated Hospital, Department of Nutrition, School of Public Health, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
27
|
Deng M, Li Z, Chen S, Wang H, Sun L, Tang J, Luo L, Zhang X, Xu H, Dai Z. Exploring the heterogeneity of hepatic and pancreatic fat deposition in obesity: implications for metabolic health. Front Endocrinol (Lausanne) 2024; 15:1447750. [PMID: 39439559 PMCID: PMC11493592 DOI: 10.3389/fendo.2024.1447750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Objective This retrospective observational study investigates the heterogeneity of hepatic and pancreatic fat deposition and its implications for metabolic health in individuals with obesity. Methods A total of 706 patients with obesity underwent an MRI to quantify liver and pancreatic fat. Patients were classified into four groups based on fat deposition: no fat (None), fatty pancreas only (NAFPD), fatty liver only (NAFLD), and both conditions (NAFLD+NAFPD). Biochemical profiles, insulin resistance (Homeostatic Model Assessment for Insulin Resistance, HOMA-IR), and β-cell function were analyzed. A series of multiple linear regressions were used to investigate the independent effects of characteristics on glucose, insulin, and C-peptide at 0h. Another multiple linear regression was performed to evaluate the effects of basic characteristics on average liver fat, mean pancreatic fat, and visceral fat. Results The majority (76.63%) exhibited both NAFLD and NAFPD, highlighting the heterogeneity of fat deposition among individuals with obesity. Groups with fatty liver displayed significantly higher fasting glucose, insulin, C-peptide, and HOMA-IR levels than those without fatty liver (P < 0.01). Fatty pancreas alone did not significantly influence these metabolic parameters (P > 0.05). This underscores the greater metabolic impact of hepatic fat compared to pancreatic fat. Conclusions The study confirms the complex heterogeneity of fat deposition in obesity, with the fatty liver being a more influential factor in metabolic disturbances than the fatty pancreas. The prevalent co-occurrence of NAFLD and NAFPD in this population underscores the need for targeted management strategies focusing on hepatic fat reduction to mitigate metabolic risk.
Collapse
Affiliation(s)
- Ming Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, Wuhan, China
| | - Zhen Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Shangyu Chen
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Endocrinology and Metabolism, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi, Nanning, China
| | - Huawei Wang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Li Sun
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jun Tang
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Liman Luo
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xiaoxiao Zhang
- Department of MSC Clinical & Technical Solutions, Philips Healthcare, Beijing, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, China
- Wuhan Clinical Research and Development Center of Brain Resuscitation and Functional Imaging, Wuhan, China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Clinical Nutrition, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Kouvari M, Valenzuela-Vallejo L, Guatibonza-Garcia V, Verrastro O, Axarloglou E, Mylonakis SC, George J, Papatheodoridis G, Mingrone G, Mantzoros CS. Apolipoprotein C-III in association with metabolic-dysfunction associated steatotic liver disease: A large, multicenter study. Clin Nutr 2024; 43:101-108. [PMID: 39442390 DOI: 10.1016/j.clnu.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND & AIMS The available literature on the effect of apolipoprotein C-III (ApoC-III) inhibition in MASLD reveals inconsistencies. The aim of the present work was to examine levels of ApoC-III in the entire spectrum of metabolic-dysfunction associated steatotic liver disease (MASLD). METHODS This is a multicenter study involving patients enrolled in two gastroenterology-hepatology clinics (Greece and Australia) and in a bariatric-metabolic surgery clinic (Italy), with liver biopsy before and after bariatric surgery or lifestyle modification. RESULTS Comparing simple MASL to steatohepatitis (MASH) with fibrosis stage F ≥ 2 (at-risk MASH), revealed a marginally significant trend for decreased ApoC-III levels in the latter group (p = 0.07). Multi-adjusted analysis revealed an inverse association between ApoC-III and at-risk MASH (Odds Ratioper 1 mg/dL increase in ApoC-III = 0.91, 95 % Confidence Interval (0.83, 0.99)). ApoC-III interacted with triglycerides in predicting at-risk MASH (p-for-interaction = 0.002). Participants with ApoC-III > median (∼3.75 mg/dL) and normal triglycerides (triglyceridese≤150 mg/dL) had the lowest likelihood to present at-risk MASH (31.8 %) in contrast with participants with ApoC-III < median and hypertriglyceridemia among whom at-risk MASH was recorded in 57.1 %. In multi-adjusted analysis participants with normal triglycerides and high ApoC-III had 64 % lower odds of at-risk MASH compared with their counterparts with ApoC-III < median (OR = 0.36, 95%CI (0.14, 0.86)). Among participants with hypertriglyceridemia, those with ApoC-III < median had less prevalent at-risk MASH compared with those with ApoC-III ≥ median (OR = 0.54, 95%CI (0.32, 0.98)); however in all cases significance was lost when liver enzymes were taken into account. CONCLUSIONS In advanced disease stages, ApoC-III levels seem to be decreased and advanced organ damage may be a potential explanation. Mendelian randomization studies are needed to confirm or refute this hypothesis.
Collapse
Affiliation(s)
- Matina Kouvari
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laura Valenzuela-Vallejo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Evangelos Axarloglou
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sophia C Mylonakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Georgios Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | | | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
29
|
Cui Y, Yang K, Guo C, Xia Z, Jiang B, Xue Y, Song B, Hu W, Zhang M, Wei Y, Zhang C, Zhang S, Fang J. Carbon monoxide as a negative feedback mechanism on HIF-1α in the progression of metabolic-associated fatty liver disease. Nitric Oxide 2024; 153:1-12. [PMID: 39369813 DOI: 10.1016/j.niox.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) encompasses various chronic liver conditions, yet lacks approved drugs. Hypoxia-inducible factor-1α (HIF-1α) is pivotal in MAFLD development. Our prior research highlighted the efficacy of the nano-designed carbon monoxide (CO) donor, targeting HIF-1α in a mouse hepatic steatosis model. Given heme oxygenase-1 (HO-1, a major downstream molecule of HIF-1α) as the primary source of intrinsic CO, we hypothesized that upregulation of HO-1/CO, responsive to HIF-1α, forms a negative feedback loop regulating MAFLD progression. In this study, we explored the potential negative feedback mechanism of CO on HIF-1α and its downstream effects on MAFLD advancement. HIF-1α emerges early in hepatic steatosis induced by a high-fat (HF) diet, triggering increased HO-1 and inflammation. SMA/CORM2 effectively suppresses HIF-1α and steatosis progression when administered within the initial week of HF diet initiation but loses impact later. In adipose tissues, concurrent metabolic dysfunction and inflammation with HIF-1α activation suggest adipose tissue expansion initiates HF-induced steatosis, triggering hypoxia and liver inflammation. Notably, in an in vitro study using mouse hepatocytes treated with fatty acids, downregulating HO-1 intensified HIF-1α induction at moderate fatty acid concentrations. However, this effect diminished at high concentrations. These results suggest the HIF-1α-HO-1-CO axis as a feedback loop under physiological and mild pathological conditions. Excessive HIF-1α upregulation in pathological conditions overwhelms the CO feedback loop. Additional CO application effectively suppresses HIF-1α and disease progression, indicating potential application for MAFLD control.
Collapse
Affiliation(s)
- Yingying Cui
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, 750000, China; Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Kai Yang
- Department of Medical Technology, Anhui Medical College, No.632, Furong Road, Hefei, Anhui Province, China
| | - Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Benchun Jiang
- Department of Gastricintestinal Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanni Xue
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Mingjie Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanyan Wei
- Department of Infectious Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Shichen Zhang
- Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China; Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto, 860-0082, Japan.
| |
Collapse
|
30
|
Nie S, Zhang S, Wang Y, Zhu M, Chen X, Wang X, Huang P. Extraction, purification, structural characterization, and bioactivities of Ginkgo biloba leave polysaccharides: A review. Int J Biol Macromol 2024; 281:136280. [PMID: 39368588 DOI: 10.1016/j.ijbiomac.2024.136280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/09/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Ginkgo biloba, a deciduous tree from the Ginkgoaceae family, is widely cultivated globally. In China, it predominantly grows in the eastern and southern regions. The leaves can be harvested multiple times throughout the growing season, presenting a significant resource potential. Ginkgo biloba leaves are considered as a living fossil with both medicinal and edible properties in traditional Chinese medicine. Polysaccharides, the primary bioactive compounds in these leaves, exhibit numerous biological activities, including antioxidant, antitumor, anti-inflammatory, immunoregulatory activity, antidepressant effects, hepatoprotective, hypoglycemic activity and hair-growth promoting effect. This review highlights the advancements in the extraction separation purification, structural elucidation, and functional analysis of polysaccharides derived from Ginkgo biloba leaves over the past decade, aiming to provide valuable insights for future development and commercialization of Ginkgo biloba leave polysaccharides.
Collapse
Affiliation(s)
- Shanshan Nie
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Shan Zhang
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yongxia Wang
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Mingjun Zhu
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xinju Chen
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xinlu Wang
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
31
|
Braha A, Timar B, Ivan V, Balica MM, Dăniluc L, Timar R. Novel Biomarkers of Grade I Left Ventricular Diastolic Dysfunction in Type 2 Diabetes Patients with Metabolic-Dysfunction-Associated Steatotic Liver Disease. J Clin Med 2024; 13:5901. [PMID: 39407960 PMCID: PMC11477181 DOI: 10.3390/jcm13195901] [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: 09/16/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Prior research has identified a significant association between heart disease and metabolic-dysfunction-associated steatotic liver disease (MASLD); however, the underlying mechanisms are unclear. This study aimed to identify predictive biomarkers associated with grade I left ventricular diastolic dysfunction (LVDD) in patients with type 2 diabetes mellitus (T2DM). Methods: This single-center, cross-sectional study evaluated 73 T2DM patients for grade 1 LVDD and MASLD using 2D echocardiography, tissue analysis, spectral color Doppler, and Fibromax. Results: This study analyzed 50 patients (mean age 58.0 ± 11.3 years) with a median diabetes duration of 7 years, abdominal obesity (mean body mass index (BMI) 34.4 ± 5.9 kg/m2), and a mean HbA1c of 7.9 ± 1.5%. The prevalence of grade I LVDD, fibrosis, mild steatosis, moderate-to-severe liver steatosis, mild MASLD, and moderate MASLD was 54%, 44%, 14%, 80%, 43%, and 34%, respectively. Regression analysis revealed that grade 1 LVDD was positively associated with age, Fibrotest, α2-macroglobulin, epicardiac adipose tissue (EAT), and negatively associated with lateral s', E wave, E/e', E/A, medium E', and septal e' (p < 0.05 for all). α2-macroglobulin > 1.92 g/L (area under the receiver operating characteristic curve (AUROC) = 0.782, sensitivity 70.4%, specificity 81.2%) and fibrotest score > 0.11 (AUROC 0.766, sensitivity 92.6%, specificity 56.2%) were significant predictors of grade I LVDD. Conclusions: Although the underlying mechanisms remain unclear, innovative non-invasive biomarkers, such as α2-macroglobulin or fibrotest, could concurrently indicate liver stiffness and the likelihood of grade I LVDD, an early, asymptomatic HF stage in T2DM patients.
Collapse
Affiliation(s)
- Adina Braha
- Department of Second Internal Medicine Diabetes, Nutrition, Metabolic Diseases and Systemic Rheumatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (R.T.)
- Department of Diabetes, Nutrition and Metabolic Diseases Clinic, “Pius Brînzeu” Emergency Clinical County University Hospital, 300723 Timisoara, Romania
| | - Bogdan Timar
- Department of Second Internal Medicine Diabetes, Nutrition, Metabolic Diseases and Systemic Rheumatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (R.T.)
- Department of Diabetes, Nutrition and Metabolic Diseases Clinic, “Pius Brînzeu” Emergency Clinical County University Hospital, 300723 Timisoara, Romania
| | - Viviana Ivan
- Second Department of Internal Medicine-Cardiology Clinic, Victor Babeş University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Cardiology Clinic, “Pius Brinzeu” Emergency County Hospital Timisoara, 300723 Timisoara, Romania
| | - Monica Micloș Balica
- Second Department of Internal Medicine-Cardiology Clinic, Victor Babeş University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Cardiology Clinic, “Pius Brinzeu” Emergency County Hospital Timisoara, 300723 Timisoara, Romania
| | - Larisa Dăniluc
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Romulus Timar
- Department of Second Internal Medicine Diabetes, Nutrition, Metabolic Diseases and Systemic Rheumatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.B.); (R.T.)
- Department of Diabetes, Nutrition and Metabolic Diseases Clinic, “Pius Brînzeu” Emergency Clinical County University Hospital, 300723 Timisoara, Romania
| |
Collapse
|
32
|
Giangregorio F, Mosconi E, Debellis MG, Provini S, Esposito C, Garolfi M, Oraka S, Kaloudi O, Mustafazade G, Marín-Baselga R, Tung-Chen Y. A Systematic Review of Metabolic Syndrome: Key Correlated Pathologies and Non-Invasive Diagnostic Approaches. J Clin Med 2024; 13:5880. [PMID: 39407941 PMCID: PMC11478146 DOI: 10.3390/jcm13195880] [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: 09/10/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background and Objectives: Metabolic syndrome (MetS) is a condition marked by a complex array of physiological, biochemical, and metabolic abnormalities, including central obesity, insulin resistance, high blood pressure, and dyslipidemia (characterized by elevated triglycerides and reduced levels of high-density lipoproteins). The pathogenesis develops from the accumulation of lipid droplets in the hepatocyte (steatosis). This accumulation, in genetically predisposed subjects and with other external stimuli (intestinal dysbiosis, high caloric diet, physical inactivity, stress), activates the production of pro-inflammatory molecules, alter autophagy, and turn on the activity of hepatic stellate cells (HSCs), provoking the low grade chronic inflammation and the fibrosis. This syndrome is associated with a significantly increased risk of developing type 2 diabetes mellitus (T2D), cardiovascular diseases (CVD), vascular, renal, pneumologic, rheumatological, sexual, cutaneous syndromes and overall mortality, with the risk rising five- to seven-fold for T2DM, three-fold for CVD, and one and a half-fold for all-cause mortality. The purpose of this narrative review is to examine metabolic syndrome as a "systemic disease" and its interaction with major internal medicine conditions such as CVD, diabetes, renal failure, and respiratory failure. It is essential for internal medicine practitioners to approach this widespread condition in a "holistic" rather than a fragmented manner, particularly in Western countries. Additionally, it is important to be aware of the non-invasive tools available for assessing this condition. Materials and Methods: We conducted an exhaustive search on PubMed up to July 2024, focusing on terms related to metabolic syndrome and other pathologies (heart, Lung (COPD, asthma, pulmonary hypertension, OSAS) and kidney failure, vascular, rheumatological (osteoarthritis, rheumatoid arthritis), endocrinological, sexual pathologies and neoplastic risks. The review was managed in accordance with the PRISMA statement. Finally, we selected 300 studies (233 papers for the first search strategy and 67 for the second one). Our review included studies that provided insights into metabolic syndrome and non-invasive techniques for evaluating liver fibrosis and steatosis. Studies that were not conducted on humans, were published in languages other than English, or did not assess changes related to heart failure were excluded. Results: The findings revealed a clear correlation between metabolic syndrome and all the pathologies above described, indicating that non-invasive assessments of hepatic fibrosis and steatosis could potentially serve as markers for the severity and progression of the diseases. Conclusions: Metabolic syndrome is a multisystem disorder that impacts organs beyond the liver and disrupts the functioning of various organs. Notably, it is linked to a higher incidence of cardiovascular diseases, independent of traditional cardiovascular risk factors. Non-invasive assessments of hepatic fibrosis and fibrosis allow clinicians to evaluate cardiovascular risk. Additionally, the ability to assess liver steatosis may open new diagnostic, therapeutic, and prognostic avenues for managing metabolic syndrome and its complications, particularly cardiovascular disease, which is the leading cause of death in these patients.
Collapse
Affiliation(s)
- Francesco Giangregorio
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Emilio Mosconi
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Maria Grazia Debellis
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Stella Provini
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Ciro Esposito
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Matteo Garolfi
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Simona Oraka
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Olga Kaloudi
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Gunel Mustafazade
- Department of Internal Medicine, Codogno Hospital, Via Marconi 1, 26900 Codogno, Italy; (F.G.); (E.M.); (M.G.D.); (S.P.); (C.E.); (M.G.); (S.O.); (G.M.)
| | - Raquel Marín-Baselga
- Department of Internal Medicine, Hospital Universitario La Paz, Paseo Castellana 241, 28046 Madrid, Spain;
| | - Yale Tung-Chen
- Department of Internal Medicine, Hospital Universitario La Paz, Paseo Castellana 241, 28046 Madrid, Spain;
| |
Collapse
|
33
|
Wu Y, Kuang Y, Wu Y, Dai H, Bi R, Hu J, Sun L. Yang-Gan-Jiang-Mei formula alleviates non-alcoholic steatohepatitis by inhibiting NLRP3 inflammasome through mitophagy. Biotechnol Genet Eng Rev 2024; 40:1314-1333. [PMID: 36960758 DOI: 10.1080/02648725.2023.2193482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
As an effective formula of traditional Chinese medicine, Yang-Gan-Jiang-Mei (YGJM) formula exhibited a unique advantage in ameliorating liver injury and hepatic steatosis of non-alcoholic steatohepatitis (NASH). Nevertheless, the related pharmacological mechanism needs to be elucidated. This study aimed to explore the molecular mechanism of YGJM formula on mitophagy mediated by PINK1/parkin signaling pathway and NOD-like receptor protein 3 (NLRP3) inflammasome in NASH. High-fat-diet rats and HepG2 cells induced by free fatty acid were used as NASH models in vivo and in vitro. Liver pathology and serum indicator embodying liver function (aspartate transferase, alanine transferase, triglyceride, and total cholesterol) were applied to evaluate the extent of hepatic damage and lipid accumulation. Besides, transmission electron microscopy, JC-1 and 2',7'-dichlorofluorescein diacetate were utilized to observe hepatic mitochondrial morphology, as well as cellular mitochondrial membrane potential and reactive oxygen species level. Additionally, expression of PINK1/parkin-mediated mitophagy and NLRP3 inflammasome was detected to elucidate the underlying mechanism of YGJM formula by immunohistochemistry, immunofluorescence, RT-PCR (reverse transcription-polymerase chain reaction) and Western blot. The manifestations of pathology and biochemical detection confirmed the efficacy of YGJM formula in relieving hepatic damage and lipid deposition. Simultaneously, YGJM formula could obviously improve mitochondrial function. In addition, YGJM formula exhibited the promotion of PINK1/parkin-mediated mitophagy, which could perturb NLRP3 inflammasome activation, and as a result, the hepatocyte inflammation was also suppressed both in vitro and in vivo. Our preliminary results indicate that YGJM formula can ameliorate NASH mechanistically by interfering with PINK1/parkin-mediated mitophagy and NLRP3 inflammasome to exert anti-inflammation ability and promote mitochondrial function restoration.
Collapse
Affiliation(s)
- Yuanyuan Wu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yufeng Kuang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunbang Wu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Heng Dai
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruiqi Bi
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaming Hu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixia Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Choudhury A, Rajaram R, Sarin SK. Acute-on-chronic liver failure in metabolic dysfunction-associated fatty liver disease patients: a disease multiplier. Hepatol Int 2024; 18:941-958. [PMID: 39107615 DOI: 10.1007/s12072-024-10711-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/30/2024] [Indexed: 10/05/2024]
Abstract
Acute-on-chronic liver failure (ACLF) is a syndrome of liver failure due to an acute hepatic insult leading to liver failure with or without extra-hepatic organ failure in a patient of chronic liver disease (CLD) with or without cirrhosis presenting for the first time. The definition is still with controversy; hence, homogeneity and clarity of the case is an unmet need. There is a paradigm shift noted as far as the etiology of CLD is concerned with rise in metabolic dysfunction-associated fatty liver disease (MAFLD) and ethanol as the dominant cause even in developing countries. MAFLD is the change in nomenclature from NAFLD to justify the metabolic derangement in these group of patients. The shift from an exclusion-based criteria to one that has evolved to a diagnosis that requires positive criteria has profound significance. Clearly there is a difference in terms of its prevalence, disease progression, and liver-related events, as well as management of metabolic risk factors and MAFLD itself which requires further understanding. In tandem with the global rise in MAFLD, the incidence of MAFLD-ACLF is increasing. Excessive alcohol consumption causes metabolic and toxic injury to the liver resulting in nearly similar pathway of fatty liver, hepatitis, and cirrhosis. The interaction of MAFLD as an additional underlying chronic liver injury in ACLF patients is complex due to the presence of metabolic risk factors that are unique to MAFLD. There is lack of clarity on how MAFLD affects the clinical course of ACLF due to scarcity of this specific data. This narrative review aims to understand the unique effects, consequences, and management of MAFLD as the chronic liver injury component in ACLF.
Collapse
Affiliation(s)
- Ashok Choudhury
- Dept of Hepatology and Liver Transplantation. Institute of Liver and Biliary Sciences, New Delhi, India
| | - Ruveena Rajaram
- Consultant, Gastroenterology and Hepatology Unit, Department of Medicine, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Shiv Kumar Sarin
- Department of Hepatology and Liver TransplantChancellor, Chancellor . Institute of Liver and Biliary Sciences, Senior Proffesor, New Delhi, 110070, India.
| |
Collapse
|
35
|
Zhang YF, Qiao W, Zhuang J, Feng H, Zhang Z, Zhang Y. Association of ultra-processed food intake with severe non-alcoholic fatty liver disease: a prospective study of 143073 UK Biobank participants. J Nutr Health Aging 2024; 28:100352. [PMID: 39340900 DOI: 10.1016/j.jnha.2024.100352] [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: 04/24/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Previous studies indicate a link between non-alcoholic fatty liver disease (NAFLD) and unhealthy dietary patterns or nutrient intake. However, it remains unclear whether ultra-processed foods (UPF) contribute to an increased risk of NAFLD. This study aimed to explore how ultra-processed food consumption correlates with severe NAFLD using the UK Biobank data. METHODS This prospective cohort study included 143,073 participants from the UK Biobank. UPF consumption levels were determined using the NOVA classification and quantified from 24-h dietary recall data. The association between UPF consumption and severe NAFLD (hospitalization or death) was initially examined using Cox proportional hazards models with intake quartiles. Nonlinear associations were investigated using penalized cubic splines fitted in the Cox proportional hazards models. Adjustments were made for general characteristics, sociodemographic factors, body mass index (BMI), and lifestyle. RESULTS Throughout the median follow-up period of 10.5 years, 1,445 participants developed severe NAFLD. The adjusted models indicated a significant increase in severe NAFLD risk in higher UPF intake groups compared to the lowest quartile (HR: 1.26 [95% CI: 1.11-1.43]). Subgroup analysis revealed that individuals with a BMI of 25 or higher were at greater risk in the highest quartile of UPF consumption. Sensitivity analyses yielded results consistent with these findings. CONCLUSION Higher consumption of UPF is associated with an increased risk of severe NAFLD. Reducing the intake of UPF can be a potential approach to lower the risk of NAFLD.
Collapse
Affiliation(s)
- Yi-Feng Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wanning Qiao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jinhong Zhuang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Hanxiao Feng
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhilan Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
36
|
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
|
37
|
Bignotto M, Bianco E, Centofanti L, Russo A, Dei Cas M, Zermiani P, Morano C, Samartin F, Bertolini E, Bifari F, Berra C, Zuin M, Paroni R, Battezzati PM, Folli F. Synergistic effects of glucose tolerance and BMI on cardiovascular events and all-cause mortality in a healthy population: CA.ME.LI.A study 7 years follow-up. Am J Physiol Endocrinol Metab 2024; 327:E498-E511. [PMID: 39196799 PMCID: PMC11482241 DOI: 10.1152/ajpendo.00181.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 08/30/2024]
Abstract
The CA.ME.LI.A (CArdiovascular risks, MEtabolic syndrome, LIver and Autoimmune disease) epidemiological study was conducted in Abbiategrasso (Milan, Italy) to identify risk factors for metabolic and cardiovascular disease in an apparently healthy population of northern Italy. The population (n = 2,545, 1,251 men, 1,254 women) was stratified according to body mass index [normal body weight (NBW): <25 kg/m2; overweight-obese (OWO): ≥25 kg/m2] and according to fasting blood glucose [normal fasting glucose: <100 mg/dL; impaired fasting glucose (IFG): 100-125 mg/dL; diabetes mellitus (DM): ≥126 mg/dL]. The incidence of cardiovascular (CV) events and overall mortality were studied by the Kaplan-Meier method using the log rank test. Univariate analysis was conducted with time-dependent Cox models. During the 7-yr follow-up period, 80 deaths and 149 CV events occurred. IFG [hazard ratio (HR): 2.81; confidence interval (CI): 1.37-5.77; P = 0.005], DM (HR: 4.88; CI: 1.47-16; P = 0.010), or OWO (HR: 2.78; CI:1.68-4.59; P < 0.001) all produced significant increases in CV events and deaths. In the combination IFG/OWO (HR: 5.51; CI: 3.34-9.08; P < 0.001), there was an apparent additive effect of the two conditions, whereas in the combination DM/OWO (HR: 12.71; CI: 7.48-22; P < 0.001), there was an apparent multiplicative effect on the risk for CV events and deaths. In males, the DM/NBW group had a higher incidence of cardiovascular events and deaths than the IFG/OWO group. In contrast, in females, the IFG/OWO group had a higher incidence of cardiovascular events and deaths than the DM/NBW group. In women, there was a greater incidence of CV events in the IFG/OWO group (HR: 6.23; CI: 2.88-13; P < 0.001) than in men in the same group (HR: 4.27; CI: 2.15-8.47; P < 0.001). Consistent with these data, also all-cause mortality was progressively increased by IFG/DM and OWO, with an apparently exponential effect in the combination DM/OWO (HR: 11.78; CI: 6.11-23; P < 0.001). IFG/DM and OWO, alone or in combination, had major effects in increasing mortality for all causes and CV events. The relative contributions of hyperglycemia and overweight/obesity on cardiovascular events and deaths were apparently, to a certain extent, sex dependent. Females were more affected by overweight/obesity either alone or combined with IFG, as compared with males.NEW & NOTEWORTHY For the first time, the combined effects of glucose tolerance and BMI have been investigated in an apparently healthy large population sample of a city in the north of Italy. We found that there are synergistic effects of glucose levels with BMI to increase not only cardiovascular events and deaths but also cancer-related deaths and all-cause mortality.
Collapse
Affiliation(s)
- Monica Bignotto
- Liver and Gastroenterology Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Bianco
- Liver and Gastroenterology Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Medicine and Liver Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Lucia Centofanti
- Clinical Biochemistry and Mass Spectrometry Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Antonio Russo
- Epidemiology Unit, Agency for Health Protection of the Metropolitan City of Milan, Milan, Italy
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Zermiani
- Liver and Gastroenterology Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Camillo Morano
- Clinical Biochemistry and Mass Spectrometry Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Federica Samartin
- Liver and Gastroenterology Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Medicine and Liver Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | | | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, LITA, Segrate, Italy
| | - Cesare Berra
- Dipartimento Endocrino-Metabolico, IRCCS MultiMedica, Milano, Italy
| | - Massimo Zuin
- Liver and Gastroenterology Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Rita Paroni
- Clinical Biochemistry and Mass Spectrometry Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Pier Maria Battezzati
- Liver and Gastroenterology Unit, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Medicine and Liver Unit, ASST Santi Paolo e Carlo, Milan, Italy
| | - Franco Folli
- Departmental Unit for Diabetes and Metabolic Diseases, ASST Santi Paolo e Carlo, Milan, Italy
- Departmental Unit for Diabetes and Metabolic Diseases, ASST Santi Paolo e Carlo, Milan, Italy
| |
Collapse
|
38
|
Crane JD, Barrandon O, Faherty B, Gorgoglione M, Crowley C, Morin J, Ross TT, Shimkonis J, Li D, Hirenallur-Shanthappa D, Boucher M, Ahn Y, Clasquin MF. Murine HSD17β13 does not control liver steatosis and modestly impacts fibrosis in a sex- and diet-specific manner. J Lipid Res 2024; 65:100634. [PMID: 39182609 PMCID: PMC11440797 DOI: 10.1016/j.jlr.2024.100634] [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: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Human genetic studies show that loss of function mutations in 17-Beta hydroxysteroid dehydrogenase (HSD17β13) are associated with protection from non-alcoholic steatohepatitis (NASH). As a result, therapies that reduce HSD17β13 are being pursued for the treatment of NASH. However, inconsistent effects on steatosis, inflammation, and fibrosis pathogenesis have been reported in murine Hsd17b13 knockdown or knockout models. To clarify whether murine Hsd17b13 loss regulates liver damage and fibrosis, we characterized Hsd17b13 knockout mice subjected to pro-NASH diets or pro-inflammatory chemical-induced liver injury. There were no effects of Hsd17b13 loss on liver injury, inflammation, fibrosis, or lipids after 28 weeks on the Gubra-Amylin NASH (GAN) diet or 12 weeks on a 45% choline-deficient high-fat diet (CDAHFD). However, AAV-mediated re-expression of murine Hsd17b13 in KO mice increased liver macrophage abundance in both sexes fed the 45% CDAHFD. In contrast, there was a modest reduction in liver fibrosis, but not lipids or inflammation within Hsd17b13 null female, but not male, mice after 12 weeks of a 60% CDAHFD compared to WT littermates. Fibrosis and the abundance of liver macrophages were increased in Hsd17b13 KO females upon adenoviral re-expression of mouse HSD17β13, but this was not reflected in inflammatory markers. Additionally, we found minimal differences in liver injury, lipids, or inflammatory and fibrotic markers 48 h after acute CCl4 exposure. In summary, murine Hsd17b13 loss has modest diet- and sex-specific effects on liver fibrosis which contrasts with human genetic studies. This suggests a disconnect between the biological function of HSD17β13 in mice and humans.
Collapse
Affiliation(s)
- Justin D Crane
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA.
| | - Ornella Barrandon
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Bryan Faherty
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Matt Gorgoglione
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Collin Crowley
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Jeff Morin
- Global Discovery Investigative and Translational Sciences CM-DSRD, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Trenton T Ross
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Jackson Shimkonis
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Dongmei Li
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | | | - Magalie Boucher
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Youngwook Ahn
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts, USA
| | | |
Collapse
|
39
|
Leow YW, Chan WL, Lai LL, Mustapha NRN, Mahadeva S, Quiambao R, Munteanu M, Chan WK. LIVERSTAT for risk stratification for patients with metabolic dysfunction-associated fatty liver disease. J Gastroenterol Hepatol 2024; 39:2182-2189. [PMID: 38946405 DOI: 10.1111/jgh.16675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/03/2024] [Accepted: 06/16/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND AND AIM LIVERSTAT is an artificial intelligence-based noninvasive test devised to screen for and provide risk stratification for metabolic dysfunction-associated fatty liver disease (MAFLD) by using simple blood biomarkers and anthropometric measurements. We aimed to study LIVERSTAT in patients with MAFLD and to explore its role for the diagnosis of advanced fibrosis. METHODS This is a retrospective study of data from MAFLD patients who underwent a liver biopsy. Patients with type 2 diabetes who underwent transient elastography and had liver stiffness measurement (LSM) < 5 kPa were included as patients with no fibrosis. Among these patients, controlled attenuation parameter <248 dB/m was considered as no steatosis. The LIVERSTAT results were generated based on a proprietary algorithm, blinded to the histological and LSM data. RESULTS The data for 350 patients were analyzed (mean age 53 years, 45% male, advanced fibrosis 22%). The sensitivity, specificity, positive predictive value, negative predictive value, and misclassification rate of LIVERSTAT to diagnose advanced fibrosis were 90%, 50%, 30%, 95%, and 42%, respectively. The corresponding rates for Fibrosis-4 score (FIB4) were 56%, 83%, 44%, 89%, and 22%, respectively. When LSM was used as a second test, the corresponding rates for LIVERSTAT were 60%, 97%, 76%, 94%, and 8%, respectively, while the corresponding rates for FIB4 were 38%, 99%, 83%, 89%, and 11%, respectively. CONCLUSION LIVERSTAT had a higher negative predictive value compared with FIB4 and a lower misclassification rate compared with FIB4 when used in a two-step approach in combination with LSM for the diagnosis of advanced fibrosis.
Collapse
Affiliation(s)
- Yong Wen Leow
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Wah Loong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lee Lee Lai
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Sanjiv Mahadeva
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ronald Quiambao
- Medical Affairs, Fibronostics US Inc., Indian Harbour Beach, Florida, USA
| | - Mona Munteanu
- Medical Affairs, Fibronostics US Inc., Indian Harbour Beach, Florida, USA
| | - Wah Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
40
|
Schophaus S, Creasy KT, Koop PH, Clusmann J, Jaeger J, Punnuru V, Koch A, Trautwein C, Loomba R, Luedde T, Schneider KM, Schneider CV. Machine learning uncovers manganese as a key nutrient associated with reduced risk of steatotic liver disease. Liver Int 2024; 44:2807-2821. [PMID: 39082383 PMCID: PMC11464189 DOI: 10.1111/liv.16055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects approximately 20%-30% of the general population and is linked to high-caloric western style diet. However, there are little data that specific nutrients might help to prevent steatosis. METHODS We analysed the UK Biobank (ID 71300) 24 h-nutritional assessments and investigated the association between nutrient intake calculated from food questionnaires and hepatic steatosis indicated by imaging or ICD10-coding. The effect of manganese (Mn) on subgroups with risk single nucleotide polymorphism carriage as well as the effect on metabolomics was investigated. All analyses are corrected for age, sex, body mass index, Townsend index for socioeconomic status, kcal, alcohol, protein intake, fat intake, carbohydrate intake, energy from beverages, diabetes, physical activity and for multiple testing. RESULTS We used a random forest classifier to analyse the feature importance of 63 nutrients and imaging-proven steatosis in a cohort of over 25 000 UK Biobank participants. Increased dietary Mn intake was associated with a lower likelihood of MRI-diagnosed steatosis. Subsequently, we conducted a cohort study in over 200 000 UK Biobank participants to explore the relationship between Mn intake and hepatic or cardiometabolic outcomes and found that higher Mn intake was associated with a lower risk of ICD-10 coded steatosis (OR = .889 [.838-.943], p < .001), independent of other potential confounders. CONCLUSION Our study provides evidence that higher Mn intake may be associated with lower odds of steatosis in a large population-based sample. These findings underline the potential role of Mn in the prevention of steatosis, but further research is needed to confirm these findings and to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Simon Schophaus
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Kate Townsend Creasy
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Institute for Translational Medicine and Therapeutics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul-Henry Koop
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Jan Clusmann
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Julius Jaeger
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Varnitha Punnuru
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander Koch
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Rohit Loomba
- Division of Gastroenterology, Department of Medicine, University of California at San Diego, San Diego, CA, USA
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Kai Markus Schneider
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Carolin V. Schneider
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
- The Institute for Translational Medicine and Therapeutics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
41
|
Bae JH. Racial and ethnic disparities in metabolic dysfunction-associated steatotic liver disease outcomes: A call for culturally sensitive interventions: Editorial 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; 30:665-668. [PMID: 39069725 DOI: 10.3350/cmh.2024.0562] [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: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024] Open
Affiliation(s)
- Jae Hyun Bae
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
42
|
Wang J, Zhao F, Brouwer LA, Buist-Homan M, Wolters JC, Moshage H, Harmsen MC. Collagen-rich liver-derived extracellular matrix hydrogels augment survival and function of primary rat liver sinusoidal endothelial cells and hepatocytes. Int J Biol Macromol 2024; 278:134717. [PMID: 39142477 DOI: 10.1016/j.ijbiomac.2024.134717] [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: 04/09/2024] [Revised: 07/11/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Liver sinusoidal endothelial cells (LSECs) are key targets for addressing metabolic dysfunction-associated steatotic liver disease (MASLD). However, isolating and culturing primary LSECs is challenging due to rapid dedifferentiation, resulting in loss of function. The extracellular matrix (ECM) likely plays a crucial role in maintaining the fate and function of LSECs. In this study, we explored the influence of liver-ECM (L-ECM) on liver cells and developed culture conditions that maintain the differentiated function of liver cells in vitro for prolonged periods. Porcine liver-derived L-ECM, containing 34.9 % protein, 0.045 % glycosaminoglycans, and negligible residual DNA (41.2 ng/mg), was utilized to culture primary rat liver cells in generated hydrogels. Proteomic analyses and molecular weight distribution of proteins of solubilized L-ECM revealed the typical diverse ECM core matrisome, with abundant collagens. L-ECM hydrogels showed suitable stiffness and stress relaxation properties. Furthermore, we demonstrated that collagen-rich L-ECM hydrogels enhanced LSECs' and hepatocytes' viability, and reduced the dedifferentiation rate of LSECs. In addition, hepatocyte function was maintained longer by culture on L-ECM hydrogels compared to traditional culturing. These beneficial effects are likely attributed to the bioactive macromolecules including collagens, and mechanical and microarchitectural properties of the L-ECM hydrogels.
Collapse
Affiliation(s)
- Junyu Wang
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands.
| | - Fenghua Zhao
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Department of Biomedical Engineering, Groningen, the Netherlands.
| | - Linda A Brouwer
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands.
| | - Manon Buist-Homan
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen, the Netherlands.
| | - Justina C Wolters
- University of Groningen, University Medical Centre Groningen, Department of Pediatrics, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Interfaculty Mass Spectrometry Center, Groningen, the Netherlands.
| | - Han Moshage
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Laboratory Medicine, Groningen, the Netherlands.
| | - Martin C Harmsen
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| |
Collapse
|
43
|
Priego-Parra BA, Triana-Romero A, Bernal-Reyes R, Icaza-Chávez ME, Martínez-Vázquez SE, Amieva-Balmori M, Cano-Contreras AD, Vivanco-Cid H, Remes-Troche JM. Comparative evaluation of APRI, FIB-4, HFS, and NFS: Scoring tools for liver fibrosis in a Mexican population with MASLD. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2024:S2255-534X(24)00075-6. [PMID: 39358200 DOI: 10.1016/j.rgmxen.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/06/2024] [Indexed: 10/04/2024]
Abstract
INTRODUCTION AND AIM Liver fibrosis is a complication of metabolic dysfunction-associated steatotic liver disease (MASLD). Given the limitations and risks of liver biopsy, examining noninvasive scoring systems that are affordable for the population is necessary. Our aim was to evaluate and compare the diagnostic yield of the APRI, FIB-4, NAFLD score, and Hepamet fibrosis score instruments for detecting liver fibrosis in Mexican subjects with MASLD. MATERIAL AND METHODS A retrospective study was conducted on a sample of subjects with MASLD. Liver fibrosis was calculated through transient liver elastography. Sociodemographic, epidemiologic, and biochemical variables were evaluated. Scores were calculated utilizing the fibrosis-4 (FIB-4) index, the aspartate aminotransaminase-to-platelet ratio index (APRI), the Hepamet fibrosis score (HFS), and the NAFLD score (NFS), and then compared. ROC curves were constructed, and the optimum cutoff points were determined utilizing the Youden index. Sensitivity, specificity, positive predictive value, negative predictive value, and likelihood ratio were calculated. RESULTS The study included 194 subjects (63% women), of whom 150 (77.3%) were classified with MASLD and 44 (22.7%) as controls with no liver disease. There was a 15.3% prevalence of advanced fibrosis. The cutoff points of 0.57 for APRI, 1.85 for FIB-4, 0.08 for HFS, and -0.058 for NFS showed diagnostic yields with areas under the ROC curves of 0.79, 0.80, 0.70, and 0.68, respectively. CONCLUSION The APRI, FIB-4, NFS, and HFS scores are useful for evaluating liver fibrosis in Mexican subjects with MASLD. Better diagnostic yield was found with the FIB-4 and APRI scores.
Collapse
Affiliation(s)
- B A Priego-Parra
- Centro de Investigaciones Biomédicas, Universidad Veracruzana, Veracruz, Mexico; Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - A Triana-Romero
- Unidad Médica de Alta Especialidad (UMAE), Hospital de Especialidades «Dr. Bernardo Sepúlveda Gutiérrez», Centro Médico Nacional Siglo XXl, Mexico City, CDMX, Mexico
| | - R Bernal-Reyes
- Grupo de Investigación MAFLD, Asociación Mexicana de Gastroenterología, Mexico City, CDMX, Mexico
| | - M E Icaza-Chávez
- Grupo de Investigación MAFLD, Asociación Mexicana de Gastroenterología, Mexico City, CDMX, Mexico
| | - S E Martínez-Vázquez
- Grupo de Investigación MAFLD, Asociación Mexicana de Gastroenterología, Mexico City, CDMX, Mexico
| | - M Amieva-Balmori
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - A D Cano-Contreras
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - H Vivanco-Cid
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico.
| | - J M Remes-Troche
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| |
Collapse
|
44
|
Alisi A, McCaughan G, Grønbæk H. Role of gut microbiota and immune cells in metabolic-associated fatty liver disease: clinical impact. Hepatol Int 2024; 18:861-872. [PMID: 38995341 DOI: 10.1007/s12072-024-10674-6] [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: 01/24/2024] [Accepted: 03/18/2024] [Indexed: 07/13/2024]
Abstract
In 2020, a revised definition of fatty liver disease associated with metabolic dysfunction (MAFLD) was proposed to replace non-alcoholic fatty liver (NAFLD). Liver steatosis and at least one of the three metabolic risk factors, including type 2 diabetes, obesity, or signs of metabolic dysregulation, are used to diagnose MAFLD. MAFLD, similarly to NAFLD, is characterized by a spectrum of disease ranging from simple steatosis to advanced metabolic steatohepatitis with or without fibrosis, and may progress to cirrhosis and liver cancer, including increased risk of other critical extrahepatic diseases. Even though the pathophysiology of MAFLD and potential therapeutic targets have been explored in great detail, there is yet no Food and Drug Administration approved treatment. Recently, gut microbiome-derived products (e.g., endotoxins and metabolites) involved in intestinal barrier disruption, systemic inflammation, and modification of intrahepatic immunity have been associated with MAFLD development and progression. Therefore, different strategies could be adopted to modify the gut microbiome to improve outcomes in early and progressive MAFLD. Here, we provide an overview of mechanisms that may link the gut microbiome and immune response during the onset of liver steatosis and progression to steatohepatitis and fibrosis in patients with MAFLD. Finally, gut microbiota-based approaches are discussed as potential personalized treatments against MAFLD.
Collapse
Affiliation(s)
- Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesu' Children Hospital, IRCCS, Rome, Italy.
| | - Geoffrey McCaughan
- A.W Morrow Gastroenterology and Liver Center, Royal Prince Alfred Hospital, Sydney, Australia
- Centenary Institute, University of Sydney, Sydney, Australia
| | - Henning Grønbæk
- Department of Hepatology & Gastroenterology, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| |
Collapse
|
45
|
Teixeira AVS, Quitete FT, Martins BC, Peixoto TC, Ribeiro MDS, Velasco PCD, Miranda C, Resende ADC, Costa DC, Atella GC, Mucci DDB, Souza-Mello V, Martins FF, Daleprane JB. Metabolic consequences of interesterified palm oil and PCB-126 co-exposure in C57BL/6 mice. Food Chem Toxicol 2024; 192:114965. [PMID: 39197524 DOI: 10.1016/j.fct.2024.114965] [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: 05/29/2024] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined as morphofunctional changes in the liver. Studies have shown that Westernized eating patterns and environmental pollutants can directly induce the development of MASLD. This study evaluates the effect of co-exposure to interesterified palm oil (IPO) and 3,3',4,4',5-pentachlorobiphenyl (PCB-126) on the progression of MASLD in an animal model. C57BL/6 mice were fed IPO and co-exposed to PCB-126 for ten weeks. The co-exposure led to an imbalance in carbohydrate metabolism, increased systemic inflammation markers, and morphofunctional changes in the liver. These liver changes included the presence of inflammatory cells, fibrosis, alterations in aspartate transaminase (AST) and alanine transaminase (ALT) enzymes, and imbalance in gene expression related to fatty acid β-oxidation, de novo lipogenesis, mitochondrial dynamics, and endoplasmic reticulum stress. Separate exposures to IPO and PCB-126 affected metabolism and MASLD progression. Nutritional and lifestyle factors may potentiate the onset and severity of MASLD.
Collapse
Affiliation(s)
- Ananda Vitoria Silva Teixeira
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Fernanda Torres Quitete
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Bruna Cadete Martins
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Thamara Cherem Peixoto
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Mayara da Silva Ribeiro
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Patricia Coelho de Velasco
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Caroline Miranda
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Angela de Castro Resende
- Laboratory of Cardiovascular Pharmacology and Medicinal Plants, Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, 20551-030, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Geórgia Correa Atella
- Medical Biochemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniela de Barros Mucci
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, 205521031, Brazil
| | - Fabiane Ferreira Martins
- Department of Morphology Federal University of Rio Grande do Norte, Rio Grande do Norte, 59078-970, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Interaction Studies between Nutrition and Genetics, Department of Basic and Experimental Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, RJ, 20550-900, Brazil.
| |
Collapse
|
46
|
Lee M, Son MJ, Hong S, Ryu J, Min J, Lee D, Lee JH, Kim ND, Park S, Kim D, Joo J, Kwak J, Kim KH, Lee Y, Keum B, Song HS, Jung Y, Kim KS, Kim G. Discovery of a selective cytochrome P450 4A inhibitor for the treatment of metabolic dysfunction-associated fatty liver disease. Clin Transl Med 2024; 14:e1816. [PMID: 39367660 PMCID: PMC11452733 DOI: 10.1002/ctm2.1816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 10/06/2024] Open
Affiliation(s)
- Minji Lee
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of Functional GenomicsKorea University of Science & Technology (UST)DaejeonRepublic of Korea
- School of PharmacySungkyunkwan UniversitySuwonRepublic of Korea
| | - Sin‐Hyoung Hong
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
| | - Jae‐Sung Ryu
- New Drug Development CenterOsong Medical Innovation FoundationCheongjuRepublic of Korea
| | - Ji‐Hyeon Min
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
- Department of Analytical Science and TechnologyGraduate School of Analytical Science and Technology (GRAST)Chungnam National UniversityDaejeonRepublic of Korea
| | - Dong‐Eon Lee
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
- Department of Bio‐Analytical ScienceUniversity of Science and Technology (UST)DaejeonRepublic of Korea
| | - Ji Hoon Lee
- New Drug Development CenterDaegu‐Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Nam Doo Kim
- New Drug Discovery & DevelopmentVORONOIBIO Inc.IncheonRepublic of Korea
| | - Shi‐Young Park
- Korea Mouse Metabolic Phenotyping CenterLee Gil Ya Cancer and Diabetes InstituteGachon UniversityIncheonSouth Korea
| | - Darong Kim
- New Drug Development CenterDaegu‐Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Jeongmin Joo
- New Drug Development CenterDaegu‐Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Jisung Kwak
- Sensor System Research CenterKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- Department of Chemical and Biological EngineeringKorea UniversitySeoulRepublic of Korea
| | - Kook Hwan Kim
- Discovery DivisionGI Innovation, Inc.SeoulSouth Korea
| | - Yong‐Ho Lee
- Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Byeong‐Rak Keum
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Hyun Seok Song
- Sensor System Research CenterKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research GroupMetropolitan Seoul CenterKorea Basic Science Institute (KBSI)SeoulRepublic of Korea
| | - Koon Soon Kim
- Division of Endocrinology and MetabolismDaejeon Endo Internal MedicineDaejeonRepublic of Korea
| | - Gun‐Hwa Kim
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
- Department of Analytical Science and TechnologyGraduate School of Analytical Science and Technology (GRAST)Chungnam National UniversityDaejeonRepublic of Korea
- Department of Bio‐Analytical ScienceUniversity of Science and Technology (UST)DaejeonRepublic of Korea
- Research and Development CenterCYPHARMADaejeonRepublic of Korea
| |
Collapse
|
47
|
Allen AM, Younossi ZM, Diehl AM, Charlton MR, Lazarus JV. Envisioning how to advance the MASH field. Nat Rev Gastroenterol Hepatol 2024; 21:726-738. [PMID: 38834817 DOI: 10.1038/s41575-024-00938-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/06/2024]
Abstract
Since 1980, the cumulative effort of scientists and health-care stakeholders has advanced the prerequisites to address metabolic dysfunction-associated steatotic liver disease (MASLD), a prevalent chronic non-communicable liver disease. This effort has led to, among others, the approval of the first drug specific for metabolic dysfunction-associated steatohepatitis (MASH; formerly known as nonalcoholic steatohepatitis). Despite substantial progress, MASLD is still a leading cause of advanced chronic liver disease, including primary liver cancer. This Perspective contextualizes the nomenclature change from nonalcoholic fatty liver disease to MASLD and proposes important considerations to accelerate further progress in the field, optimize patient-centric multidisciplinary care pathways, advance pharmacological, behavioural and diagnostic research, and address health disparities. Key regulatory and other steps necessary to optimize the approval and access to upcoming additional pharmacological therapeutic agents for MASH are also outlined. We conclude by calling for increased education and awareness, enhanced health system preparedness, and concerted action by policy-makers to further the public health and policy agenda to achieve at least parity with other non-communicable diseases and to aid in growing the community of practice to reduce the human and economic burden and end the public health threat of MASLD and MASH by 2030.
Collapse
Affiliation(s)
- Alina M Allen
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zobair M Younossi
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, VA, USA
- The Global NASH Council, Washington DC, USA
| | | | - Michael R Charlton
- Center for Liver Diseases, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jeffrey V Lazarus
- The Global NASH Council, Washington DC, USA.
- CUNY Graduate School of Public Health and Health Policy (CUNY SPH), New York, NY, USA.
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain.
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
48
|
Ktenopoulos N, Sagris M, Gerogianni M, Pamporis K, Apostolos A, Balampanis K, Tsioufis K, Toutouzas K, Tousoulis D. Non-Alcoholic Fatty Liver Disease and Coronary Artery Disease: A Bidirectional Association Based on Endothelial Dysfunction. Int J Mol Sci 2024; 25:10595. [PMID: 39408924 PMCID: PMC11477211 DOI: 10.3390/ijms251910595] [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: 08/24/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease and is regarded as a liver manifestation of metabolic syndrome. It is linked to insulin resistance, obesity, and diabetes mellitus, all of which increase the risk of cardiovascular complications. Endothelial dysfunction (EnD) constitutes the main driver in the progression of atherosclerosis and coronary artery disease (CAD). Several pathophysiological alterations and molecular mechanisms are involved in the development of EnD in patients with NAFLD. Our aim is to examine the association of NAFLD and CAD with the parallel assessment of EnD, discussing the pathophysiological mechanisms and the genetic background that underpin this relationship. This review delves into the management of the condition, exploring potential clinical implications and available medical treatment options to facilitate the deployment of optimal treatment strategies for these patients.
Collapse
Affiliation(s)
- Nikolaos Ktenopoulos
- First Department of Cardiology, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.S.); (A.A.); (K.T.); (K.T.); (D.T.)
| | - Marios Sagris
- First Department of Cardiology, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.S.); (A.A.); (K.T.); (K.T.); (D.T.)
| | - Maria Gerogianni
- Endocrine Unit, 2nd Propaedeutic Department of Internal Medicine, School of Medicine, Research Institute and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, 12641 Athens, Greece;
- Second Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Konstantinos Pamporis
- Department of Hygiene, Social-Preventive Medicine & Medical Statistics, Medical School, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece;
| | - Anastasios Apostolos
- First Department of Cardiology, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.S.); (A.A.); (K.T.); (K.T.); (D.T.)
| | - Konstantinos Balampanis
- Second Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Konstantinos Tsioufis
- First Department of Cardiology, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.S.); (A.A.); (K.T.); (K.T.); (D.T.)
| | - Konstantinos Toutouzas
- First Department of Cardiology, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.S.); (A.A.); (K.T.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- First Department of Cardiology, ‘Hippokration’ General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.S.); (A.A.); (K.T.); (K.T.); (D.T.)
| |
Collapse
|
49
|
Wei X, Wu D, Li J, Wu M, Li Q, Che Z, Cheng X, Cheng Q, Yin F, Zhang H, Wang X, Abtahi S, Zuo L, Hang L, Ma L, Kuo WT, Liu X, Turner JR, Wang H, Xiao J, Wang F. Myeloid beta-arrestin 2 depletion attenuates metabolic dysfunction-associated steatohepatitis via the metabolic reprogramming of macrophages. Cell Metab 2024; 36:2281-2297.e7. [PMID: 39305895 DOI: 10.1016/j.cmet.2024.08.010] [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: 02/01/2023] [Revised: 05/27/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024]
Abstract
Macrophage-mediated inflammation has been implicated in the pathogenesis of metabolic dysfunction-associated steatohepatitis (MASH); however, the immunometabolic program underlying the regulation of macrophage activation remains unclear. Beta-arrestin 2, a multifunctional adaptor protein, is highly expressed in bone marrow tissues and macrophages and is involved in metabolism disorders. Here, we observed that β-arrestin 2 expression was significantly increased in the liver macrophages and circulating monocytes of patients with MASH compared with healthy controls and positively correlated with the severity of metabolic dysfunction-associated steatotic liver disease (MASLD). Global or myeloid Arrb2 deficiency prevented the development of MASH in mice. Further study showed that β-arrestin 2 acted as an adaptor protein and promoted ubiquitination of immune responsive gene 1 (IRG1) to prevent increased itaconate production in macrophages, which resulted in enhanced succinate dehydrogenase activity, thereby promoting the release of mitochondrial reactive oxygen species and M1 polarization. Myeloid β-arrestin 2 depletion may be a potential approach for MASH.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dongqing Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Li
- College of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Miaomiao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; College of Pharmacy, Anhui Medical University, Hefei, China
| | - Qianhui Li
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xu Cheng
- Department of Cardiology, First Affiliated Hospital, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Qianying Cheng
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fan Yin
- Department of Pharmacy, Huainan First People's Hospital, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan, China
| | - Hao Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuefu Wang
- College of Pharmacy, Anhui Medical University, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Li Zuo
- School of Basic Medical Sciences, Molecular Biology Laboratory, Anhui Medical University, Hefei, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Lei Hang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Wei-Ting Kuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Xiaoying Liu
- College of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China; Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Fei Wang
- Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
50
|
Song BG, Kim A, Goh MJ, Kang W, Gwak GY, Paik YH, Choi MS, Lee JH, Sinn DH. Risk of Hepatocellular Carcinoma by Steatotic Liver Disease and Its Newly Proposed Subclassification. Liver Cancer 2024; 13:561-571. [PMID: 39435269 PMCID: PMC11493391 DOI: 10.1159/000538301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/06/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Steatotic liver disease (SLD) is a new overarching term proposed to replace nonalcoholic fatty liver disease and metabolic dysfunction-associated fatty liver disease. Subclassification includes metabolic dysfunction-associated SLD (MASLD), MASLD with increased alcohol intake (MetALD), and cryptogenic SLD. This study aimed to investigate whether SLD and its subclassification could stratify hepatocellular carcinoma (HCC) risk. Methods A cohort of 85,119 adults without viral hepatitis or heavy alcohol intake was analyzed for the risk of HCC according to SLD and its subclassification. The fibrosis-4 (FIB-4) index was used to estimate the degree of liver fibrosis. Results During a median follow-up of 11.9 years, HCC was diagnosed in 123 individuals. The incidence rate of HCC per 1,000 person-years was higher in individuals with SLD than in those without SLD (0.197 vs. 0.071, p < 0.001), with an adjusted hazard ratio of 2.02 (95% confidence interval: 1.40-2.92). The HCC incidence rate per 1,000 person-years was 0, 0.180, and 0.648 for cryptogenic SLD, MASLD, and MetALD, respectively. When participants with SLD was further stratified by the FIB-4 index, the HCC incidence rate per 1,000 person-years was 0.074 for SLD with FIB-4 < 1.3 and 0.673 for SLD with FIB-4 ≥ 1.3. Of note, HCC risk was substantially high (HCC incidence rate: 1.847 per 1,000 person-years) for MetALD with FIB-4 ≥ 1.3. Conclusions HCC risk was different by SLD and its subclassification. The utilization of SLD and its subclassification can aid in stratifying HCC risk and facilitate the identification of individuals requiring interventions to mitigate the risk of HCC.
Collapse
Affiliation(s)
- Byeong Geun Song
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Aryoung Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Myung Ji Goh
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Wonseok Kang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Geum-Youn Gwak
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong-Han Paik
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Moon Seok Choi
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Hyeok Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Dong Hyun Sinn
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|