1
|
Kumar A, Arora A, Sharma P, Jan S, Ara I. Visceral Fat and Diabetes: Associations With Liver Fibrosis in Metabolic Dysfunction-Associated Steatotic Liver Disease. J Clin Exp Hepatol 2025; 15:102378. [PMID: 39268479 PMCID: PMC11387673 DOI: 10.1016/j.jceh.2024.102378] [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: 06/05/2023] [Accepted: 07/16/2024] [Indexed: 09/15/2024] Open
Abstract
Background The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease, is increasing globally. Noninvasive methods, such as bioelectrical impedance analysis (BIA), which measures body composition, including visceral fat, are gaining interest in evaluating MASLD patients. Our study aimed to identify factors associated with significant liver fibrosis, compare noninvasive scores, and highlight the importance of visceral fat measurement using BIA. Methods MASLD patients seen in our out-patient department underwent comprehensive evaluations, including liver stiffness using transient elastography, body composition analysis using BIA, and metabolic measurements. Significant fibrosis was defined as a liver stiffness measurement of ≥8.2 kPa. Using multivariate analysis, we identified factors associated with significant liver fibrosis and compared four noninvasive scores with a novel diabetes-visceral fat 15 (DVF15) score. Results We analyzed data from 609 MASLD patients seen between February 2022 and March 2023. The median age was 43 years (81% male). Among these, 78 (13%) had significant fibrosis. Patients with significant fibrosis had higher rates of type 2 diabetes (41% vs 21%, P < 0.001) and elevated levels of aspartate aminotransferase, alanine aminotransferase, hemoglobin A1c, Fibosis-4, aspartate-aminotransferase-to platelet-ratio index, and NAFLD fibrosis scores. They also exhibited higher visceral and subcutaneous fat. Binary logistic regression revealed type 2 diabetes and a visceral fat level of >15% as associated with significant liver fibrosis. Additionally, the DVF15 score, combining these factors, showed a modest area under the receiver operating characteristic curve of 0.664 (P < 0.001). Conclusion Our study identified diabetes and high visceral fat as factors associated with significant liver fibrosis in MASLD patients. We recommend that visceral fat measurement using BIA be an essential part of MASLD evaluation. The presence of either diabetes or a visceral fat level of >15% should prompt clinicians to check for significant fibrosis in MASLD patients. Further research is warranted to validate our findings and evaluate the utility of the DVF15 score in larger cohorts and diverse populations.
Collapse
Affiliation(s)
- Ashish Kumar
- Institute of Liver, Gastroenterology, & Pancreatico-Biliary Sciences, Sir Ganga Ram Hospital, New Delhi, India
| | - Anil Arora
- Institute of Liver, Gastroenterology, & Pancreatico-Biliary Sciences, Sir Ganga Ram Hospital, New Delhi, India
| | - Praveen Sharma
- Institute of Liver, Gastroenterology, & Pancreatico-Biliary Sciences, Sir Ganga Ram Hospital, New Delhi, India
| | - Shayesta Jan
- Institute of Liver, Gastroenterology, & Pancreatico-Biliary Sciences, Sir Ganga Ram Hospital, New Delhi, India
| | - Ishrat Ara
- Institute of Liver, Gastroenterology, & Pancreatico-Biliary Sciences, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
2
|
ElSayed NA, McCoy RG, Aleppo G, Bajaj M, Balapattabi K, Beverly EA, Briggs Early K, Bruemmer D, Cusi K, Echouffo-Tcheugui JB, Ekhlaspour L, Fleming TK, Garg R, Khunti K, Lal R, Levin SR, Lingvay I, Matfin G, Napoli N, Pandya N, Parish SJ, Pekas EJ, Pilla SJ, Pirih FQ, Polsky S, Segal AR, Jeffrie Seley J, Stanton RC, Verduzco-Gutierrez M, Younossi ZM, Bannuru RR. 4. Comprehensive Medical Evaluation and Assessment of Comorbidities: Standards of Care in Diabetes-2025. Diabetes Care 2025; 48:S59-S85. [PMID: 39651988 PMCID: PMC11635044 DOI: 10.2337/dc25-s004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
|
3
|
Yu H, Su X, Tao W, Sun W, Zhang X, Han Q, Zhao Z, Zhang Y, Chen X, Liu X, Jia D, Fang L, Li L. Prevalence and characteristics of liver steatosis and fibrosis in type 2 diabetes mellitus (T2DM) patients: a cross-sectional study in populations of eastern China. BMJ Open 2024; 14:e087550. [PMID: 39672583 DOI: 10.1136/bmjopen-2024-087550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2024] Open
Abstract
OBJECTIVES To describe the prevalence, clinical characteristics and risk factors of liver steatosis and fibrosis in type 2 diabetes mellitus (T2DM) patients in eastern China. DESIGN A cross-sectional, multicentre study based on an ongoing cohort study. SETTING 16 clinics in eastern China, including primary clinics to tertiary hospitals. PARTICIPANTS 1816 patients with T2DM diagnosis who met the inclusion criteria were recruited into the study. INTERVENTION Participants underwent elastography examination. MAIN OUTCOME MEASURES Descriptive analysis was performed to calculate the prevalence and characteristics of liver steatosis and fibrosis. The correlated factors were analysed using single- and multivariate logistic regression analysis. RESULTS The prevalence of liver steatosis in T2DM patients is 69.7%, with 46% moderate to severe steatosis. 34.6% and 6.7% of the patients were detected with liver fibrosis and cirrhosis. Steatosis patients were younger, had higher body mass index (BMI), higher levels of insulin resistance and more severe lipid metabolism disorders. Similar trends of differences were observed in patients with fibrosis. Female gender (OR=0.574, 95% CI 0.381 to 0.865), BMI (OR=1.491, 95% CI 1.375 to 1.616), disease duration, inflammation and serum lipid profile markers were risk factors of steatosis, while BMI (OR=1.204, 95% CI 1.137 to 1.275) and female gender (OR=0.672, 95% CI 0.470 to 0.961) were still the most significant predictors of liver fibrosis. CONCLUSIONS The prevalence of liver steatosis and fibrosis were high in patients with T2DM. Liver steatosis and fibrosis in these patients appeared to be more associated with lipid metabolism disorders and insulin resistance rather than glucose levels. TRIAL REGISTRATION NUMBER Clinical trial: NCT05597709.
Collapse
Affiliation(s)
- Hekai Yu
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
- Southeast University, Nanjing, Jiangsu, China
| | - Xianghui Su
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, Xinjiang, China
| | - Wenxuan Tao
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
- Southeast University, Nanjing, Jiangsu, China
| | - Weixia Sun
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
- Southeast University, Nanjing, Jiangsu, China
| | - Xiaoyan Zhang
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
| | - Qing Han
- Department of Endocrinology, Southeast University, Nanjing, Jiangsu, China
| | - Zhuoxiao Zhao
- Nanjing Gaochun Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Yan Zhang
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaoqian Chen
- Department of Endocrinology, Nanjing Central Hospital, Nanjing, Jiangsu, China
| | - Xinliang Liu
- Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Dianrong Jia
- Department of Endocrinology, Taizhou Jiangyan Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu, China
| | - Li Fang
- Nanjing Gaochun Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Ling Li
- Department of Endocrinology, School of Medicine, Southeast University Zhongda Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Lindfors A, Strandberg R, Hagström H. Screening for advanced liver fibrosis due to metabolic dysfunction-associated steatotic liver disease alongside retina scanning in people with type 2 diabetes: a cross-sectional study. Lancet Gastroenterol Hepatol 2024:S2468-1253(24)00313-3. [PMID: 39675369 DOI: 10.1016/s2468-1253(24)00313-3] [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] [Received: 07/03/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND International guidelines suggest screening for advanced fibrosis due to metabolic dysfunction-associated steatotic liver disease in people with type 2 diabetes, but how to implement these guidelines in clinical care remains unclear. We hypothesise that examination with VCTE could be implemented simultaneously with retina scanning with a high acceptance rate in people with type 2 diabetes. METHODS In this cross-sectional study, we offered VCTE to people with type 2 diabetes referred to routine retina scanning in a large retina scanning facility in Stockholm, Sweden. We excluded people with type 1 diabetes, currently pregnant, with known liver disease, reporting high alcohol consumption, who did not speak Swedish, or younger than 18 years. Between Nov 6, 2020, and June 20, 2023, we conducted surveys with included participants and collected data from medical records on diabetes retinopathy, sex, and VCTE measurements. Increased liver stiffness was defined as at least 8·0 kPa, and possible advanced fibrosis as more than 12·0 kPa. Presence of metabolic dysfunction-associated steatotic liver disease was defined as a controlled attenuation parameter (CAP) value of 280 dB/m or higher. Participants with a liver stiffness measurement of at least 8·0 kPa or those with unreliable measurements were subsequently referred for a secondary evaluation at a liver specialist, including a follow-up liver stiffness measurement with VCTE. The primary outcome was the proportion of eligible people approached for screening who accepted. Secondary outcomes were the prevalence of elevated liver stiffness (≥8·0 kPa or >12·0 kPa), presence of metabolic dysfunction-associated steatotic liver disease, and the proportion of elevated liver stiffness readings at the first VCTE examination that were not elevated in the secondary evaluation with a liver specialist. Secondary outcomes were assessed in all participants who accepted screening, except false positives, which were assessed only in participants who had a second examination. FINDINGS 1301 participants were eligible to undergo assessment with VCTE, which was accepted by 1005 (77·2%). 973 (96·8%) participants had complete measurements, of whom 504 (51·8%) had CAP values of 280 dB/m or higher, indicating metabolic dysfunction-associated steatotic liver disease. Of 977 participants with reliable liver stiffness measurements, 154 (15·8%) had values of at least 8·0 kPa, suggestive of liver fibrosis, and 49 (5·0%) had values higher than 12·0 kPa, indicating possible advanced fibrosis. However, upon reassessment with a second VCTE after referral, 56 (45·2%) of 124 individuals had values less than 8·0 kPa. 74 (7·4%) of 1005 participants had a final liver stiffness of at least 8·0 kPa; 29 (2·9%) had values greater than 12·0 kPa. INTERPRETATION Simultaneous examination with VCTE alongside retina scanning had a high acceptance rate among people with type 2 diabetes and could be a strategy for case-finding of people with fibrosis due to metabolic dysfunction-associated steatotic liver disease. However, a high proportion of participants in our study with elevated liver stiffness measurement at the screening visit did not have an elevated liver stiffness measurement at secondary evaluation, suggesting false-positive findings were common. FUNDING Gilead Sciences, Pfizer, Region Stockholm, Åke Wiberg Foundation, and Bengt Ihre Foundation.
Collapse
Affiliation(s)
- Andrea Lindfors
- Division of Hepatology, Department of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Hannes Hagström
- Division of Hepatology, Department of Upper Gastrointestinal Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
5
|
Selvarajah J, Raj AS. Time to move beyond FIB-4 first for MASLD screening in Type 2 diabetes. Liver Int 2024; 44:3283. [PMID: 39327752 DOI: 10.1111/liv.16057] [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: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Janakan Selvarajah
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ashok S Raj
- Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Anastasiou G, Stefanakis K, Hill MA, Mantzoros CS. Expanding diagnostic and therapeutic horizons for MASH: Comparison of the latest and conventional therapeutic approaches. Metabolism 2024; 161:156044. [PMID: 39362519 DOI: 10.1016/j.metabol.2024.156044] [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: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Affiliation(s)
- Georgia Anastasiou
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Konstantinos Stefanakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael A Hill
- Dalton Cardiovascular Research Center and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Putri RR, Casswall T, Danielsson P, Marcus C, Hagman E. Steatotic Liver Disease in Pediatric Obesity and Increased Risk for Youth-Onset Type 2 Diabetes. Diabetes Care 2024; 47:2196-2204. [PMID: 39373987 DOI: 10.2337/dc24-1236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE To assess 1) the association between metabolic dysfunction-associated steatotic liver disease (MASLD) in pediatric obesity and youth-onset type 2 diabetes, 2) the joint effect of MASLD and intermediate hyperglycemia on type 2 diabetes risk, and 3) the effect of obesity treatment on type 2 diabetes risk. RESEARCH DESIGN AND METHODS A cohort study using the Swedish Childhood Obesity Treatment Register (Barnobesitas Registret i Sverige [BORIS]) (1999-2020) linked with national registers was conducted. We included 10,346 children with overweight or obesity and 59,336 matched control individuals. MASLD was defined by transaminases and diagnosis code, separately. Type 2 diabetes was ascertained from national registers. RESULTS In the obesity cohort, median age at type 2 diabetes diagnosis was 16.9 (quartile 1 [Q1], quartile 3 [Q3]: 14.7, 21.4) years, median follow-up was 8.1 (Q1, Q3: 5.1, 11.7) years. Cumulative incidence of type 2 diabetes at age 30 was 22.7% (obesity and MASLD), 9.9% (obesity alone), and 0.7% (control individuals). MASLD was associated with risk for type 2 diabetes (hazard ratio [HR] 2.71 [95% CI 2.14-3.43]), independently of age, sex, degree of obesity, intermediate hyperglycemia, and parental type 2 diabetes. Joint effect of MASLD and intermediate hyperglycemia increased type 2 diabetes risk (HR 9.04 [6.38-12.79]). Optimal response in obesity treatment reduced the risk (HR 0.23 [0.09-0.57]). CONCLUSIONS MASLD, defined by transaminases or diagnosis code, in pediatric obesity is associated with increased risk for youth-onset type 2 diabetes. MASLD interacts synergistically with intermediate hyperglycemia to dramatically increase the risk. Optimal response in obesity treatment reduces type 2 diabetes risk, despite MASLD.
Collapse
Affiliation(s)
- Resthie R Putri
- Division of Pediatrics, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Thomas Casswall
- Division of Pediatrics, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Pernilla Danielsson
- Division of Pediatrics, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Claude Marcus
- Division of Pediatrics, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Emilia Hagman
- Division of Pediatrics, Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Angelini G, Russo S, Carli F, Infelise P, Panunzi S, Bertuzzi A, Caristo ME, Lembo E, Calce R, Bornstein SR, Gastaldelli A, Mingrone G. Dodecanedioic acid prevents and reverses metabolic-associated liver disease and obesity and ameliorates liver fibrosis in a rodent model of diet-induced obesity. FASEB J 2024; 38:e70202. [PMID: 39600104 PMCID: PMC11599784 DOI: 10.1096/fj.202402108r] [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: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Dodecanedioic acid (DC12) is a dicarboxylic acid present in protective polymers of fruit and leaves. We explored the effects of DC12 on metabolic dysfunction-associated steatohepatitis (MASH) and obesity. DC12 supplementation (100 mg/kg/day) was added to a high-fat diet (HFD) for 8 weeks in rodents to assess its impact on obesity and MASH prevention. Rats given DC12 experienced significant reductions of weight gain, liver and visceral fat weight, and improved glucose tolerance and insulin sensitivity. Liver histology showed protection against diet-induced MASH, with reduced steatosis, hepatocyte ballooning, and fibrosis. For weight-loss and MASH reversion, rats were fed HFD for 14 weeks, followed by 6 weeks with or without DC12. DC12 supplementation (100 mg/kg/day) led to a significant reduction of weight gain and liver weight. DC12 induced white adipose tissue beiging and reduced adiposity with a decrease of visceral fat. It also improved glucose tolerance, insulin sensitivity, and reduced hepatic gluconeogenic gene expression. Liver histology revealed a significant reduction in steatosis, hepatocyte ballooning, and inflammation as well as fibrosis, indicating MASH reversal. DC12 reduced hepatic lipogenesis enzymes as well as de novo lipogenesis measured by deuterated water and increased fatty acid β-oxidation. Plasma lipid profile showed lower triglycerides and phosphatidylcholines in the DC12 group. Notably, DC12 decreased mINDY expression, the cell membrane Na+-coupled citrate transporter, reducing citrate uptake and de-novo lipogenesis, linking its effects to improved lipid metabolism and reduced steatosis. We found that during the hepatic first pass, half of the DC12 ingested with water was taken up by the liver. The concentration of DC12 in the portal vein falls within the range identified in vitro as sufficient to inhibit citrate transport in hepatocytes.
Collapse
Affiliation(s)
- Giulia Angelini
- Department of Translational Medicine and SurgeryUniversità Cattolica del Sacro CuoreRomeItaly
- Department of Medical and Surgical SciencesFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
| | - Sara Russo
- Department of Translational Medicine and SurgeryUniversità Cattolica del Sacro CuoreRomeItaly
- Department of Medical and Surgical SciencesFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
| | - Fabrizia Carli
- Cardiometabolic Risk LaboratoryInstitute of Clinical Physiology (IFC), National Research Council (CNR)PisaItaly
| | - Patrizia Infelise
- Cardiometabolic Risk LaboratoryInstitute of Clinical Physiology (IFC), National Research Council (CNR)PisaItaly
| | - Simona Panunzi
- CNR‐IASI, Laboratorio di Biomatematica, Consiglio Nazionale delle RicercheIstituto di Analisi dei Sistemi ed InformaticaRomeItaly
| | - Alessandro Bertuzzi
- CNR‐IASI, Consiglio Nazionale delle RicercheIstituto di Analisi dei Sistemi ed Informatica, Laboratorio di BiomatematicaRomeItaly
| | - Maria Emiliana Caristo
- Department of Translational Medicine and SurgeryUniversità Cattolica del Sacro CuoreRomeItaly
| | - Erminia Lembo
- Department of Translational Medicine and SurgeryUniversità Cattolica del Sacro CuoreRomeItaly
- Department of Medical and Surgical SciencesFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
| | - Roberta Calce
- Department of Translational Medicine and SurgeryUniversità Cattolica del Sacro CuoreRomeItaly
- Department of Medical and Surgical SciencesFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
| | - Stefan R. Bornstein
- Department of Medicine IIIUniversitätsklinikum Carl Gustav Carus an der Technischen Universität DresdenDresdenGermany
- Division of Diabetes & Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| | - Amalia Gastaldelli
- Cardiometabolic Risk LaboratoryInstitute of Clinical Physiology (IFC), National Research Council (CNR)PisaItaly
| | - Geltrude Mingrone
- Department of Translational Medicine and SurgeryUniversità Cattolica del Sacro CuoreRomeItaly
- Department of Medical and Surgical SciencesFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
- Division of Diabetes & Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonUK
| |
Collapse
|
9
|
Huang N, Su X, Yu T, Wu X, Lu B, Sun W, Yao L, Wang M, Wang Y, Wu W, Liu Y, Yang T, Gao R, Miao C, Li L. Serum 25-hydroxy vitamin D level is associated with elastography-detected liver fibrosis in patients with type 2 diabetes mellitus in China. Front Endocrinol (Lausanne) 2024; 15:1420088. [PMID: 39698035 PMCID: PMC11653015 DOI: 10.3389/fendo.2024.1420088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024] Open
Abstract
Objective In this cross-sectional study including patients with type 2 diabetes mellitus (T2DM) we aimed to explore the relationship between serum 25-hydroxy vitamin (25(OH)D) level and liver steatosis and fibrosis in the Chinese population. Methods Patients visiting 16 clinical centers with T2DM were recruited. Their liver steatosis and fibrosis status were then assessed using elastography. Factors associated with steatosis and fibrosis were explored using regression analysis. Correlations between serum 25(OH)D levels and other patient characteristics were analyzed using linear regression. Results In total, 1,513 patients with T2DM were included in the study. The prevalence of steatosis and fibrosis was 69.7%, and 34.6%, separately. A lower level of 25(OH)D was detected in patients with liver steatosis compared to those without, although it was not an independent predictor of this condition. However, 25(OH)D level was independently associated with liver fibrosis even when adjusted for age, sex, body mass index, hemoglobin A1c, insulin, and homeostatic model assessment of insulin resistance (OR = 0.964 [0.935-0.993], P = 0.015). When patients were separated into subgroups by sex, a correlation between 25(OH)D and fibrosis was identified in the male group (OR = 0.969 [0.940-0.998], P = 0.038). Conclusions In conclusion, this multi-center, cross-sectional study in patients with T2DM showed that serum 25-hydroxy vitamin D level was strongly associated with liver fibrosis and this relationship was more pronounced in male patients. Clinical trial registration https://clinicaltrials.gov/ct2/show/, identifier NCT05597709.
Collapse
Affiliation(s)
- Nan Huang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Xianghui Su
- Department of Endocrinology, Changji Branch, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Ting Yu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiaodong Wu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Bing Lu
- Department of Endocrinology, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, China
| | - Weixia Sun
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| | - Liqin Yao
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Maoyun Wang
- Department of Endocrinology, Baoying Hospital of Traditional Chinese Medicine, Yangzhou, China
| | - Yao Wang
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Wenxuan Wu
- Department of Endocrinology, Yixing Hospital of Traditional Chinese Medicine, Yixing, China
| | - Yingzhao Liu
- Department of Endocrinology, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Ting Yang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ruidong Gao
- Department of Endocrinology, Baoying Hospital of Traditional Chinese Medicine, Yangzhou, China
| | - Congqing Miao
- Department of Endocrinology, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Institute of Glucose and Lipid Metabolism, Southeast University, Nanjing, China
| |
Collapse
|
10
|
Lee HA, Lee HA, Kim HY. Evolution of characteristics of MASLD with and without diabetes: a meta-analysis of placebo arms. Sci Rep 2024; 14:28951. [PMID: 39578601 PMCID: PMC11584620 DOI: 10.1038/s41598-024-79428-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND We explored the changes in metabolic dysfunction-associated steatotic liver disease (MASLD) severity over time by analyzing data from the placebo arms of randomized controlled trials (RCTs), focusing on the presence of diabetes. METHODS RCTs on MASLD that included a placebo arm were identified using a systematic search of the literature. Primary outcomes were changes in hepatic steatosis and fibrosis. RESULTS The meta-analysis included 8 RCTs involving 386 patients without diabetes and 24 RCTs involving 637 patients with diabetes. The pooled estimate of mean change in steatosis grade was - 0.1 in patients without diabetes, and - 0.37 in patients with diabetes (P = 0.066). The mean change in fibrosis stage was 0.05 in patients without diabetes, and - 0.03 in patients with diabetes (P = 0.359). The mean change in nonalcoholic fatty liver disease activity score was - 0.55 in patients without diabetes, and - 1.50 in patients with diabetes (P = 0.100). The mean change in ALT and AST were significantly larger in patients without diabetes compared to those with diabetes (P < 0.05). CONCLUSION Placebo treatment had a greater effect in improving liver steatosis in patients with diabetes compared to those without. These findings highlight the importance of tailored treatment strategies in MASLD, particularly considering diabetes status.
Collapse
Affiliation(s)
- Han Ah Lee
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hye Ah Lee
- Clinical Trial Center, Ewha Womans University College of Medicine, Seoul, Korea
| | - Hwi Young Kim
- Department of Internal Medicine, College of Medicine, Ewha Womans University, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, 07985, Korea.
| |
Collapse
|
11
|
Sabatini S, Sen P, Carli F, Pezzica S, Rosso C, Lembo E, Verrastro O, Daly A, Govaere O, Cockell S, Hyötyläinen T, Mingrone G, Bugianesi E, Anstee QM, Orešič M, Gastaldelli A. Hepatic glucose production rises with the histological severity of metabolic dysfunction-associated steatohepatitis. Cell Rep Med 2024; 5:101820. [PMID: 39566466 PMCID: PMC11604487 DOI: 10.1016/j.xcrm.2024.101820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 07/25/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and steatohepatitis (MASH) are associated with a high prevalence of type 2 diabetes (T2D). Individuals with MASLD exhibit insulin resistance (IR) and hyperglycemia, but it is unclear whether hepatic glucose production (HGP) is increased with MASLD severity. We evaluated HGP in a cohort of histologically characterized individuals with MASL/MASH using stable isotope infusion (6,6-2H2-glucose, U-2H5-glycerol) and liver-specific genome-scale metabolic models (GEMs). Tracer-measured HGP is increased with liver fibrosis and inflammation, but not steatosis, and is associated with lipolysis and IR. The GEM-derived gluconeogenesis is elevated due to high glucogenic/energy metabolite uptakes (lactate, glycerol, and free fatty acid [FFA]), and the expression of insulin action genes (IRS1, IRS2, and AKT2) is reduced in MASH with fibrosis F2-F4, with/without T2D, suggesting these as putative mechanisms for increased fasting HGP and hyperglycemia. In conclusion, elevated HGP, lipolysis, and IR help to explain the mechanisms for the increased risk of hyperglycemia and T2D in MASH.
Collapse
Affiliation(s)
- Silvia Sabatini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, 56121 Pisa, Italy
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Fabrizia Carli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, 56121 Pisa, Italy
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, 56121 Pisa, Italy
| | - Chiara Rosso
- Department of Medical Sciences, Division of Gastro-Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, 10124 Turin, Italy
| | - Erminia Lembo
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ornella Verrastro
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ann Daly
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Olivier Govaere
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Department of Imaging and Pathology, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Simon Cockell
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Tuulia Hyötyläinen
- School of Science and Technology, Örebro University, 70281 Örebro, Sweden
| | - Geltrude Mingrone
- Department of Medical and Surgical Sciences, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Division of Diabetes & Nutritional Sciences, School of Cardiovascular and Metabolic Medicine & Sciences, King's College Hospital, London, UK
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastro-Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, 10124 Turin, Italy
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; School of Medical Sciences, Örebro University, 70281 Örebro, Sweden.
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, 56121 Pisa, Italy; Diabetes Division, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
12
|
Motiño O, Lambertucci F, Joseph A, Durand S, Anagnostopoulos G, Li S, Carbonnier V, Nogueira-Recalde U, Montégut L, Chen H, Aprahamian F, Nirmalathasan N, Maiuri MC, Pietrocola F, Valla D, Laouénan C, Gautier JF, Castera L, Martins I, Kroemer G. ACBP/DBI neutralization for the experimental treatment of fatty liver disease. Cell Death Differ 2024:10.1038/s41418-024-01410-6. [PMID: 39550516 DOI: 10.1038/s41418-024-01410-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
Acyl-CoA binding protein (ACBP), also known as diazepam-binding inhibitor (DBI), is an extracellular checkpoint of autophagy. Here, we report that patients with histologically confirmed metabolic-associated steatohepatitis (MASH) or liver fibrosis exhibit elevated levels of circulating ACBP/DBI protein as compared to non-affected controls. Plasma ACBP/DBI strongly correlated with the NAFLD and FIB4 scores in patients, and these correlations were independent of age and body mass index. We studied the capacity of a monoclonal antibody (mAb) neutralizing mouse ACBP/DBI to combat active liver disease in several mouse models, in which steatohepatitis had been induced by four different protocols, namely, (i) methionine/choline-deficient diet, (ii) Western style diet (WD) alone, (iii) WD combined with the hepatotoxic agent CCl4, and (iv) a combination of CCl4 injections and oral ethanol challenge. Injections of anti-ACBP/DBI mAb attenuated histological, enzymological, metabolomic and transcriptomic signs of liver damage in these four models, hence halting or reducing the progression of non-alcoholic and alcoholic liver disease. Steatosis, inflammation, ballooning and fibrosis responded to ACBP/DBI inhibition at the preclinical level. Altogether, these findings support a causal role of ACBP/DBI in MASH and liver fibrosis, as well as the possibility to therapeutically target ACBP/DBI.
Collapse
Affiliation(s)
- Omar Motiño
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Unidad de Excelencia, Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - CSIC, Valladolid, Spain.
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Adrien Joseph
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Gerasimos Anagnostopoulos
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sijing Li
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Uxía Nogueira-Recalde
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Fundación Profesor Novoa Santos, A Coruña, Spain
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Fanny Aprahamian
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Nitharsshini Nirmalathasan
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Naples, Italy
| | - Federico Pietrocola
- Department of Bioscience and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Dominique Valla
- Université Paris Cité, UMR1149 (CRI), Inserm, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Cédric Laouénan
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm U1137, Laboratory "Infection, Antimicrobials, Modelling, Evolution" (IAME), Paris, France
- Département d'Epidémiologie Biostatistique et Recherche Clinique, AP-HP.Nord, Hôpital Bichat, Paris, France
| | - Jean-François Gautier
- Institut Necker Enfants Malades, Inserm U1151, CNRS UMR 8253, IMMEDIAB Laboratory, Paris, France
- Centre Universitaire de Diabétologie et de ses Complications, AP-HP, Hôpital Lariboisiére, Paris, France
| | - Laurent Castera
- Université Paris Cité, UMR1149 (CRI), Inserm, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Inserm U1138, Université de Paris, Sorbonne Université, Equipe labellisée par la Ligue contre le cancer, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Institut Universitaire de France, Paris, France.
| |
Collapse
|
13
|
Gancheva S, Roden M, Castera L. Diabetes as a risk factor for MASH progression. Diabetes Res Clin Pract 2024; 217:111846. [PMID: 39245423 DOI: 10.1016/j.diabres.2024.111846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Non-alcoholic (now: metabolic) steatohepatitis (MASH) is the progressive inflammatory form of metabolic dysfunction-associated steatotic liver disease (MASLD), which often coexists and mutually interacts with type 2 diabetes (T2D), resulting in worse hepatic and cardiovascular outcomes. Understanding the intricate mechanisms of diabetes-related MASH progression is crucial for effective therapeutic strategies. This review delineates the multifaceted pathways involved in this interplay and explores potential therapeutic implications. The synergy between adipose tissue, gut microbiota, and hepatic alterations plays a pivotal role in disease progression. Adipose tissue dysfunction, particularly in the visceral depot, coupled with dysbiosis in the gut microbiota, exacerbates hepatic injury and insulin resistance. Hepatic lipid accumulation, oxidative stress, and endoplasmic reticulum stress further potentiate inflammation and fibrosis, contributing to disease severity. Dietary modification with weight reduction and exercise prove crucial in managing T2D-related MASH. Additionally, various well-known but also novel anti-hyperglycemic medications exhibit potential in reducing liver lipid content and, in some cases, improving MASH histology. Therapies targeting incretin receptors show promise in managing T2D-related MASH, while thyroid hormone receptor-β agonism has proven effective as a treatment of MASH and fibrosis.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich-Heine University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany.
| | - Laurent Castera
- Department of Hepatology, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France; Université Paris-Cité, INSERM UMR 1149, Centre de Recherche sur l'Inflammation Paris, Montmartre, Paris, France.
| |
Collapse
|
14
|
Colosimo S, Miller H, Koutoukidis DA, Marjot T, Tan GD, Harman DJ, Aithal GP, Manousou P, Forlano R, Parker R, Sheridan DA, Newsome PN, Alazawi W, Cobbold JF, Tomlinson JW. Glycated haemoglobin is a major predictor of disease severity in patients with NAFLD. Diabetes Res Clin Pract 2024; 217:111820. [PMID: 39147101 DOI: 10.1016/j.diabres.2024.111820] [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: 01/18/2024] [Revised: 06/30/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES Currently, non-invasive scoring systems to stage the severity of non-alcoholic fatty liver disease (NAFLD) do not consider markers of glucose control (glycated haemoglobin, HbA1c); this study aimed to define the relationship between HbA1c and NAFLD severity in patients with and without type 2 diabetes. RESEARCH DESIGN AND METHODS Data were obtained from 857 patients with liver biopsy staged NAFLD. Generalized-linear models and binomial regression analysis were used to define the relationships between histological NAFLD severity, age, HbA1c, and BMI. Paired biopsies from interventional studies (n = 421) were used to assess the impact of change in weight, HbA1c and active vs. placebo treatment on improvements in steatosis, non-alcoholic steatohepatitis (NASH), and fibrosis. RESULTS In the discovery cohort (n = 687), risk of severe steatosis, NASH and advanced fibrosis correlated positively with HbA1c, after adjustment for obesity and age. These data were endorsed in a separate validation cohort (n = 170). Predictive modelling using HbA1c and age was non-inferior to the established non-invasive biomarker, Fib-4, and allowed the generation of HbA1c, age, and BMI adjusted risk charts to predict NAFLD severity. Following intervention, reduction in HbA1c was associated with improvements in steatosis and NASH after adjustment for weight change and treatment, whilst fibrosis change was only associated with weight change and treatment. CONCLUSIONS HbA1c is highly informative in predicting NAFLD severity and contributes more than BMI. Assessments of HbA1c must be a fundamental part of the holistic assessment of patients with NAFLD and, alongside age, can be used to identify patients with highest risk of advanced disease.
Collapse
Affiliation(s)
- Santo Colosimo
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Hamish Miller
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Barts Liver Centre, Queen Mary University London and Barts Health NHS Trust, London, UK
| | - Dimitrios A Koutoukidis
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford Liver Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; UK NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Garry D Tan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - David J Harman
- Royal Berkshire Hospital NHS Foundation Trust, Reading, UK
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Pinelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Richard Parker
- Leeds Liver Unit, St James's University Hospital Leeds, Leeds, UK
| | - David A Sheridan
- Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - Philip N Newsome
- National Institute for Health Research Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK
| | - William Alazawi
- Barts Liver Centre, Queen Mary University London and Barts Health NHS Trust, London, UK; Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jeremy F Cobbold
- Oxford Liver Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; UK NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
15
|
Gracen L, Aikebuse M, Sarraf B, McPhail SM, Russell AW, O'Beirne J, Irvine KM, Williams S, Valery PC, Powell EE. An Australian Community-Based Metabolic Dysfunction-Associated Steatotic Liver Disease Care Pathway for People with Type 2 Diabetes: Barriers and Considerations. Patient Prefer Adherence 2024; 18:1845-1855. [PMID: 39280346 PMCID: PMC11397175 DOI: 10.2147/ppa.s468705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
Background Although clinical guidelines endorse screening for metabolic dysfunction-associated steatotic liver disease (MASLD) with advanced fibrosis in people with type 2 diabetes (T2D), the feasibility of and barriers and considerations relevant to implementing this approach in the community remain unclear. Methods Sequential adults with T2D attending selected community clinics during 2021-2023 were invited to receive a "liver health check" (n=543). A further 95 participants were referred directly from their general practitioner (GP) or self-referred to the study. A total of 302 participants underwent a point of care assessment of hepatic steatosis and stiffness (FibroScan) and were advised to see their GP to discuss the results. "Template" letters containing key results, their interpretation and advice about management of cardiometabolic risk, patient follow-up and referral criteria, were sent to participants' GPs. Results Referral to a tertiary liver clinic was advised in GP letters for 45 (15%) participants with an increased risk of clinically significant fibrosis (liver stiffness measurement ≥8), 15 participants with 'red flags' (eg splenomegaly, thrombocytopenia) and 2 with unsuccessful FibroScan examinations. A referral from GPs to the liver clinic was received for 27 (44%) of these 62 participants. Approximately 90% of GPs rated the "template" letters favourably on a Likert rating scale. Conclusion The low rate of participation in the "liver health check" and liver clinic referral reflects a real-world scenario and may stem from societal under-recognition and engagement with MASLD, competing health priorities or under-appreciation of the link between liver fibrosis severity and mortality risk. Further studies need to address strategies to enhance participation in liver health assessments and determine their impact on liver-related morbidity/mortality and overall survival.
Collapse
Affiliation(s)
- Lucy Gracen
- Department of Gastroenterology and Hepatology, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Melanie Aikebuse
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
- Centre for Liver Disease Research, Faculty of Medicine, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Babak Sarraf
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
- Centre for Liver Disease Research, Faculty of Medicine, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Steven M McPhail
- Australian Centre for Health Services Innovation School of Public Health, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Anthony W Russell
- Endocrinology and Diabetes, the Alfred Hospital, Melbourne, VIC, Australia
| | - James O'Beirne
- Department of Gastroenterology and Hepatology, Sunshine Coast University Hospital, Birtinya, QLD, Australia
| | - Katharine M Irvine
- Mater Research, Translational Research Institute, Brisbane, QLD, Australia
| | | | | | - Elizabeth E Powell
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
- Centre for Liver Disease Research, Faculty of Medicine, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
16
|
Tacke F, Horn P, Wai-Sun Wong V, Ratziu V, Bugianesi E, Francque S, Zelber-Sagi S, Valenti L, Roden M, Schick F, Yki-Järvinen H, Gastaldelli A, Vettor R, Frühbeck G, Dicker D. EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol 2024; 81:492-542. [PMID: 38851997 DOI: 10.1016/j.jhep.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 06/10/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
17
|
Suwała S, Junik R. Assessment of the Liver Steatosis and Fibrosis Risk in Metabolic Syndrome and Its Individual Components, Considering the Varying Definitions Used in Clinical Practice throughout Time: A Retrospective Cross-Sectional Study. Biomedicines 2024; 12:1739. [PMID: 39200204 PMCID: PMC11351204 DOI: 10.3390/biomedicines12081739] [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: 07/06/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Multiple modifications of metabolic syndrome diagnostic criteria have been made-NCEP: ATP III (from 2001, modified in 2004), IDF (2005), IDF Consortium (2009), or Polish Scientific Society Consortium standards (2022) are now frequently in use. Hepatosteatosis and hepatofibrosis are commonly mentioned aspects of metabolic syndrome that greatly increase the likelihood of developing complications. The objective of the study was to assess different diagnostic criteria for metabolic syndrome based on the prevalence of liver steatosis and fibrosis. A retrospective analysis was conducted on the medical data of 2102 patients. Out of all the single criteria, meeting the obesity criterion based on waist circumference showed the highest increase in the risk of hepatosteatosis (by 64-69%, depending on the definition used)-hypertriglyceridemia increased the risk of hepatofibrosis by 71%. Regardless of the specific criteria used, patients with metabolic syndrome had a 34-36% increased likelihood of developing hepatosteatosis-the probability of hepatofibrosis varied between 42% and 47% for the criteria established in 2004, 2005, and 2009, while the Polish 2022 criteria were not statistically significant (p = 0.818). It seems appropriate to establish consistent metabolic syndrome diagnostic criteria-the 2009 IDF guidelines are the most effective in assessing hepatosteatosis and fibrosis risk.
Collapse
Affiliation(s)
- Szymon Suwała
- Department of Endocrinology and Diabetology, Nicolaus Copernicus University, Collegium Medicum, 9 Sklodowskiej-Curie Street, 85-094 Bydgoszcz, Poland;
| | | |
Collapse
|
18
|
Jeong BK, Choi WI, Choi W, Moon J, Lee WH, Choi C, Choi IY, Lee SH, Kim JK, Ju YS, Kim P, Moon YA, Park JY, Kim H. A male mouse model for metabolic dysfunction-associated steatotic liver disease and hepatocellular carcinoma. Nat Commun 2024; 15:6506. [PMID: 39090079 PMCID: PMC11294468 DOI: 10.1038/s41467-024-50660-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 07/16/2024] [Indexed: 08/04/2024] Open
Abstract
The lack of an appropriate preclinical model of metabolic dysfunction-associated steatotic liver disease (MASLD) that recapitulates the whole disease spectrum impedes exploration of disease pathophysiology and the development of effective treatment strategies. Here, we develop a mouse model (Streptozotocin with high-fat diet, STZ + HFD) that gradually develops fatty liver, metabolic dysfunction-associated steatohepatitis (MASH), hepatic fibrosis, and hepatocellular carcinoma (HCC) in the context of metabolic dysfunction. The hepatic transcriptomic features of STZ + HFD mice closely reflect those of patients with obesity accompanying type 2 diabetes mellitus, MASH, and MASLD-related HCC. Dietary changes and tirzepatide administration alleviate MASH, hepatic fibrosis, and hepatic tumorigenesis in STZ + HFD mice. In conclusion, a murine model recapitulating the main histopathologic, transcriptomic, and metabolic alterations observed in MASLD patients is successfully established.
Collapse
Affiliation(s)
- Byung-Kwan Jeong
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
- Biomedical Research Center, KAIST, Daejeon, Korea
| | - Won-Il Choi
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
- Biomedical Research Center, KAIST, Daejeon, Korea
| | - Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea.
| | - Jieun Moon
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
- Biomedical Research Center, KAIST, Daejeon, Korea
| | - Won Hee Lee
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
- Biomedical Research Center, KAIST, Daejeon, Korea
| | - Chan Choi
- Department of Pathology, Chonnam National University Medical School, Hwasun, Korea
| | - In Young Choi
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd, Hwaseong, Korea
| | - Sang-Hyun Lee
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd, Hwaseong, Korea
| | - Jung Kuk Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co. Ltd, Hwaseong, Korea
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
- Biomedical Research Center, KAIST, Daejeon, Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea
- Biomedical Research Center, KAIST, Daejeon, Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, 22212, Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Liver Center, Severance Hospital, Seoul, Korea.
| | - Hail Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Korea.
- Biomedical Research Center, KAIST, Daejeon, Korea.
| |
Collapse
|
19
|
Castera L, Garteiser P, Laouenan C, Vidal-Trécan T, Vallet-Pichard A, Manchon P, Paradis V, Czernichow S, Roulot D, Larger E, Pol S, Bedossa P, Correas JM, Valla D, Gautier JF, Van Beers BE. Prospective head-to-head comparison of non-invasive scores for diagnosis of fibrotic MASH in patients with type 2 diabetes. J Hepatol 2024; 81:195-206. [PMID: 38548067 DOI: 10.1016/j.jhep.2024.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND & AIMS Non-invasive scores have been proposed to identify patients with fibrotic, metabolic dysfunction-associated steatohepatitis (MASH), who are at the highest risk of progression to complications of cirrhosis and may benefit from pharmacologic treatments. However, data in patients with type 2 diabetes (T2DM) are lacking. The aim of this multicenter prospective study was to perform a head-to-head comparison of FAST (FibroScan-aspartate aminotransferase [AST]), MAST (MRI-AST), MEFIB (magnetic resonance elastography [MRE] plus FIB-4), and FNI (fibrotic NASH index) for detecting fibrotic MASH in patients with T2DM. METHODS A total of 330 outpatients with T2DM and biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD) from the QUID-NASH study (NCT03634098), who underwent FibroScan, MRI-proton density fat fraction and MRE at the time of liver biopsy were studied. The main outcome was fibrotic MASH, defined as NAS ≥4 (with at least one point for each parameter) and fibrosis stage ≥2 (centrally reviewed). RESULTS All data for score comparisons were available for 245 patients (median age 59 years, 65% male, median BMI 31 kg/m2; fibrotic MASH in 39%). FAST and MAST had similar accuracy (AUROCs 0.81 vs. 0.79, p = 0.41) but outperformed FNI (0.74; p = 0.01) and MEFIB (0.68; p <0.0001). When using original cut-offs, MAST outperformed FAST, MEFIB and FNI when comparing the percentage of correctly classified patients, in whom liver biopsy would be avoided (69% vs. 48%, 46%, 39%, respectively; p <0.001). When using cut-offs specific to our population, FAST outperformed FNI and MAST (56% vs. 40%, and 38%, respectively; p <0.001). CONCLUSION Our findings show that FAST, MAST, MEFIB and FNI are accurate non-invasive tools to identify patients with T2DM and fibrotic MASH in secondary/tertiary diabetes clinics. Cut-offs adapted to the T2DM population should be considered. IMPACT AND IMPLICATIONS Among patients with type 2 diabetes (T2DM), identifying those with metabolic dysfunction-associated steatohepatitis and significant fibrosis, who are the most at risk of developing clinical liver-related outcomes and who may benefit from pharmacologic treatments, is an unmet need. In this prospective multicenter study, we compared four non-invasive scores, three based on imaging (MRI or ultrasound technologies) and one on laboratory blood tests, for this purpose, using original and study-specific cut-offs. Our findings show that FAST, MAST, MEFIB and FNI are accurate non-invasive tools to identify patients with T2DM and fibrotic MASH in secondary/tertiary diabetes clinics. Cut-offs adapted to the T2DM population should be considered. TRIAL REGISTRATION NUMBER NCT03634098.
Collapse
Affiliation(s)
- Laurent Castera
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'hépatologie, AP-HP, Hôpital Beaujon, F-92110 Clichy-la-Garenne, France.
| | | | - Cédric Laouenan
- Université Paris Cité, UMR1137 (IAME), Inserm, F-75018 Paris, France; (DEBRC), APHP, Hôpital Bichat, Paris, France
| | - Tiphaine Vidal-Trécan
- Centre Universitaire du Diabète et de ses Complications, AP-HP, Hôpital Lariboisière, F-75010 Paris, France
| | | | | | - Valérie Paradis
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'anatomie et de cytologie pathologiques, AP-HP, Hôpital Beaujon, 792110 Clichy-la-Garenne, France
| | - Sébastien Czernichow
- INSERM UMR-S1151, CNRS UMR-S8253, Immediab lab, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France; Service de nutrition, centre spécialisé Obésité, APHP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Dominique Roulot
- Université Paris-Est, U955, Inserm, F-94000 Créteil, France; Unité d'hépatologie, AP-HP, Hôpital Avicenne, 93000 Bobigny, France
| | - Etienne Larger
- Université Paris Cité, Institut Cochin, U1016, Inserm, F-75014 Paris, France; Service de diabétologie, AP-HP, Groupe hospitalier Cochin, F-75014 Paris, France
| | - Stanislas Pol
- Service d'hépatologie, AP-HP, Groupe hospitalier Cochin, F-75014 Paris, France; Université Paris Cité, Institut Cochin, U1016, Inserm, F-75014 Paris, France
| | - Pierre Bedossa
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Liverpat, F-75116 Paris, France
| | - Jean-Michel Correas
- Sorbonne Université, CNRS, INSERM Laboratoire d'Imagerie Biomédicale, Paris, France; Service d'Imagerie Adulte, AP-HP, Hôpital Necker Enfants Malades, F-75015 Paris, France
| | - Dominique Valla
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'hépatologie, AP-HP, Hôpital Beaujon, F-92110 Clichy-la-Garenne, France
| | - Jean-François Gautier
- Centre Universitaire du Diabète et de ses Complications, AP-HP, Hôpital Lariboisière, F-75010 Paris, France; INSERM UMR-S1151, CNRS UMR-S8253, Immediab lab, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Bernard E Van Beers
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service de Radiologie, AP-HP, Hôpital Beaujon, F-92110 Clichy-la-Garenne, France
| |
Collapse
|
20
|
Selvarajah J, Fourlanos S, Raj A. Noninvasive Fibrosis Tests Require Further Evaluation in At-risk Populations. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00682-7. [PMID: 39074524 DOI: 10.1016/j.cgh.2024.07.011] [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: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024]
Affiliation(s)
- Janakan Selvarajah
- Department of Gastroenterology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Spiros Fourlanos
- Department of Endocrinology, Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Ashok Raj
- Department of Gastroenterology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Yang X, Qiu K, Jiang Y, Huang Y, Zhang Y, Liao Y. Metabolic Crosstalk between Liver and Brain: From Diseases to Mechanisms. Int J Mol Sci 2024; 25:7621. [PMID: 39062868 PMCID: PMC11277155 DOI: 10.3390/ijms25147621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Multiple organs and tissues coordinate to respond to dietary and environmental challenges. It is interorgan crosstalk that contributes to systemic metabolic homeostasis. The liver and brain, as key metabolic organs, have their unique dialogue to transmit metabolic messages. The interconnected pathogenesis of liver and brain is implicated in numerous metabolic and neurodegenerative disorders. Recent insights have positioned the liver not only as a central metabolic hub but also as an endocrine organ, capable of secreting hepatokines that transmit metabolic signals throughout the body via the bloodstream. Metabolites from the liver or gut microbiota also facilitate a complex dialogue between liver and brain. In parallel to humoral factors, the neural pathways, particularly the hypothalamic nuclei and autonomic nervous system, are pivotal in modulating the bilateral metabolic interplay between the cerebral and hepatic compartments. The term "liver-brain axis" vividly portrays this interaction. At the end of this review, we summarize cutting-edge technical advancements that have enabled the observation and manipulation of these signals, including genetic engineering, molecular tracing, and delivery technologies. These innovations are paving the way for a deeper understanding of the liver-brain axis and its role in metabolic homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunfei Liao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
22
|
Barrera F, Uribe J, Olvares N, Huerta P, Cabrera D, Romero-Gómez M. The Janus of a disease: Diabetes and metabolic dysfunction-associated fatty liver disease. Ann Hepatol 2024; 29:101501. [PMID: 38631419 DOI: 10.1016/j.aohep.2024.101501] [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: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2024]
Abstract
Metabolic Dysfunction-Associated Fatty Liver Disease and Diabetes Mellitus are two prevalent metabolic disorders that often coexist and synergistically contribute to the progression of each other. Several pathophysiological pathways are involved in the association, including insulin resistance, inflammation, and lipotoxicity, providing a foundation for understanding the complex interrelationships between these conditions. The presence of MASLD has a significant impact on diabetes risk and the development of microvascular and macrovascular complications, and diabetes significantly contributes to an increased risk of liver fibrosis progression in MASLD and the development of hepatocellular carcinoma. Moreover, both pathologies have a synergistic effect on cardiovascular events and mortality. Therapeutic interventions targeting MASLD and diabetes are discussed, considering lifestyle modifications, pharmacological agents, and emerging treatment modalities. The review also addresses the challenges in managing these comorbidities, such as the need for personalized approaches and the potential impact on cardiovascular health. The insights gleaned from this analysis can inform clinicians, researchers, and policymakers in developing integrated strategies for preventing, diagnosing, and managing these metabolic disorders.
Collapse
Affiliation(s)
- Francisco Barrera
- Laboratorio Experimental de Hepatología, Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Javier Uribe
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nixa Olvares
- Laboratorio Experimental de Hepatología, Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Programa de Immunogenética e Inmunología traslacional, Instituto de Ciencias e Inovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Paula Huerta
- Programa de Medicina Interna, Instituto de Ciencias e Inovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile; Hospital Padre Hurtado, Santiago, Chile
| | - Daniel Cabrera
- Laboratorio Experimental de Hepatología, Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; Escuela de Medicina, Facultad de Ciencias Médicas, Universidad Bernardo O Higgins, Santiago, Chile
| | - Manuel Romero-Gómez
- Enfermedades Digestivas y Ciberehd, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (CSIC/HUVR/US), Universidad de Sevilla, Sevilla, España.
| |
Collapse
|
23
|
Pramanik S, Pal P, Ray S. Non-alcoholic fatty liver disease in type 2 diabetes: Emerging evidence of benefit of peroxisome proliferator-activated receptors agonists and incretin-based therapies. World J Methodol 2024; 14:91319. [PMID: 38983664 PMCID: PMC11229880 DOI: 10.5662/wjm.v14.i2.91319] [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: 12/27/2023] [Revised: 01/20/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global epidemic, affecting more than half of the people living with type 2 diabetes (T2D). The relationship between NAFLD and T2D is bidirectional and the presence of one perpetuates the other, which significantly increases the hepatic as well as extrahepatic complications. Until recently, there was no approved pharmacological treatment for NAFLD/ nonalcoholic steatohepatitits (NASH). However, there is evidence that drugs used for diabetes may have beneficial effects on NAFLD. Insulin sensitizers acting through peroxisome proliferator-activated receptor (PPAR) modulation act on multiple levels of NAFLD pathogenesis. Pioglitazone (PPARγ agonist) and saroglitazar (PPARα/γ agonist) are particularly beneficial and recommended by several authoritative bodies for treating NAFLD in T2D, although data on biopsy-proven NASH are lacking with the latter. Initial data on elafibanor (PPAR α/δ agonist) and Lanifibranor (pan PPAR agonist) are promising. On the other hand, incretin therapies based on glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RA) and dual- and triple-hormone receptor co-agonists reported impressive weight loss and may have anti-inflammatory and antifibrotic properties. GLP-1 RAs have shown beneficial effects on NAFLD/NASH and more studies on potential direct effects on liver function by dual- and triple-agonists are required. Furthermore, the long-term safety of these therapies in NAFLD needs to be established. Collaborative efforts among healthcare providers such as primary care doctors, hepatologists, and endocrinologists are warranted for selecting patients for the best possible management of NAFLD in T2D.
Collapse
Affiliation(s)
- Subhodip Pramanik
- Department of Endocrinology, Neotia Getwel Multispecialty Hospital, Siliguri 734010, West Bengal, India
| | - Partha Pal
- Department of Medical Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India
| | - Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar 751019, Odisha, India
| |
Collapse
|
24
|
Huang CX, Zhou XD, Pan CQ, Zheng MH. Screening for metabolic dysfunction-associated fatty liver disease: Time to discard the emperor's clothes of normal liver enzymes? World J Gastroenterol 2024; 30:2839-2842. [PMID: 38947289 PMCID: PMC11212714 DOI: 10.3748/wjg.v30.i22.2839] [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: 03/10/2024] [Revised: 04/28/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most prevalent chronic liver condition worldwide. Current liver enzyme-based screening methods have limitations that may missed diagnoses and treatment delays. Regarding Chen et al, the risk of developing MAFLD remains elevated even when alanine aminotransferase levels fall within the normal range. Therefore, there is an urgent need for advanced diagnostic techniques and updated algorithms to enhance the accuracy of MAFLD diagnosis and enable early intervention. This paper proposes two potential screening methods for identifying individuals who may be at risk of developing MAFLD: Lowering these thresholds and promoting the use of noninvasive liver fibrosis scores.
Collapse
Affiliation(s)
- Chen-Xiao Huang
- Metabolic Dysfunction-Associated Fatty Liver Disease Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Xiao-Dong Zhou
- Department of Cardiovascular Medicine, The Key Laboratory of Cardiovascular Diseases of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Calvin Q Pan
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York University Grossman School of Medicine, New York, NY 11355, United States
| | - Ming-Hua Zheng
- Metabolic Dysfunction-Associated Fatty Liver Disease Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
- Institute of Hepatology, Wenzhou Medical University, Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
25
|
Sahashi Y, Vukadinovic M, Amrollahi F, Trivedi H, Rhee J, Chen J, Cheng S, Ouyang D, Kwan AC. Opportunistic Screening of Chronic Liver Disease with Deep Learning Enhanced Echocardiography. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.13.24308898. [PMID: 38947008 PMCID: PMC11213089 DOI: 10.1101/2024.06.13.24308898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Importance Chronic liver disease affects more than 1.5 billion adults worldwide, however the majority of cases are asymptomatic and undiagnosed. Echocardiography is broadly performed and visualizes the liver; but this information is not leveraged. Objective To develop and evaluate a deep learning algorithm on echocardiography videos to enable opportunistic screening for chronic liver disease. Design Retrospective observational cohorts. Setting Two large urban academic medical centers. Participants Adult patients who received echocardiography and abdominal imaging (either abdominal ultrasound or abdominal magnetic resonance imaging) with ≤30 days between tests, between July 4, 2012, to June 4, 2022. Exposure Deep learning model predictions from a deep-learning computer vision pipeline that identifies subcostal view echocardiogram videos and detects the presence of cirrhosis or steatotic liver disease (SLD). Main Outcome and Measures Clinical diagnosis by paired abdominal ultrasound or magnetic resonance imaging (MRI). Results A total of 1,596,640 echocardiogram videos (66,922 studies from 24,276 patients) from Cedars-Sinai Medical Center (CSMC) were used to develop EchoNet-Liver, an automated pipeline that identifies high quality subcostal images from echocardiogram studies and detects the presence of cirrhosis or SLD. In the held-out CSMC test cohort, EchoNet-Liver was able to detect the presence of cirrhosis with an AUC of 0.837 (0.789 - 0.880) and SLD with an AUC of 0.799 (0.758 - 0.837). In a separate test cohort with paired abdominal MRIs, cirrhosis was detected with an AUC of 0.704 (0.689-0.718) and SLD was detected with an AUC of 0.726 (0.659-0.790). In an external test cohort of 106 patients (n = 5,280 videos), the model detected cirrhosis with an AUC of 0.830 (0.738 - 0.909) and SLD with an AUC of 0.768 (0.652 - 0.875). Conclusions and Relevance Deep learning assessment of clinical echocardiography enables opportunistic screening of SLD and cirrhosis. Application of this algorithm may identify patients who may benefit from further diagnostic testing and treatment for chronic liver disease.
Collapse
Affiliation(s)
- Yuki Sahashi
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Milos Vukadinovic
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA
| | - Fatemeh Amrollahi
- Bioinformatics Research, Department of Medicine, Stanford University, Palo Alto, CA
| | - Hirsh Trivedi
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Justin Rhee
- School of Medicine, Brown University, Providence, RI
| | - Jonathan Chen
- Bioinformatics Research, Department of Medicine, Stanford University, Palo Alto, CA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - David Ouyang
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Division of Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Alan C Kwan
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
26
|
EASL-EASD-EASO Clinical Practice Guidelines on the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Obes Facts 2024; 17:374-444. [PMID: 38852583 PMCID: PMC11299976 DOI: 10.1159/000539371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
27
|
Solleiro-Villavicencio H, Méndez-García LA, Ocampo-Aguilera NA, Baltazar-Pérez I, Arreola-Miranda JA, Aguayo-Guerrero JA, Alfaro-Cruz A, González-Chávez A, Fonseca-Sánchez MA, Fragoso JM, Escobedo G. Decreased Hepatic and Serum Levels of IL-10 Concur with Increased Lobular Inflammation in Morbidly Obese Patients. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:862. [PMID: 38929479 PMCID: PMC11205754 DOI: 10.3390/medicina60060862] [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: 04/26/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and ranges from simple steatosis to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. Accumulating evidence in animal models suggests that loss of interleukin-10 (IL-10) anti-inflammatory actions might contribute to lobular inflammation, considered one of the first steps toward NASH development. However, the role of IL-10 in lobular inflammation remains poorly explored in humans. We examined mRNA and protein levels of IL-10 in liver biopsies and serum samples from morbidly obese patients, investigating the relationship between IL-10 and lobular inflammation degree. Materials and Methods: We prospectively enrolled morbidly obese patients of both sexes, assessing the lobular inflammation grade by the Brunt scoring system to categorize participants into mild (n = 7), moderate (n = 19), or severe (n = 13) lobular inflammation groups. We quantified the hepatic mRNA expression of IL-10 by quantitative polymerase chain reaction and protein IL-10 levels in liver and serum samples by Luminex Assay. We estimated statistical differences by one-way analysis of variance (ANOVA) and Tukey's multiple comparison test. Results: The hepatic expression of IL-10 significantly diminished in patients with severe lobular inflammation compared with the moderate lobular inflammation group (p = 0.01). The hepatic IL-10 protein levels decreased in patients with moderate or severe lobular inflammation compared with the mild lobular inflammation group (p = 0.008 and p = 0.0008, respectively). In circulation, IL-10 also significantly decreased in subjects with moderate or severe lobular inflammation compared with the mild lobular inflammation group (p = 0.005 and p < 0.0001, respectively). Conclusions: In liver biopsies and serum samples of morbidly obese patients, the protein levels of IL-10 progressively decrease as lobular inflammation increases, supporting the hypothesis that lobular inflammation develops because of the loss of the IL-10-mediated anti-inflammatory counterbalance.
Collapse
Affiliation(s)
| | - Lucía Angélica Méndez-García
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Nydia A. Ocampo-Aguilera
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Itzel Baltazar-Pérez
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - José A. Arreola-Miranda
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - José A. Aguayo-Guerrero
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Ana Alfaro-Cruz
- Pathological Anatomy Department, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | - Antonio González-Chávez
- Clínica de Atención Integral para Pacientes con Diabetes y Obesidad (CAIDO), General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | | | - José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| | - Galileo Escobedo
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| |
Collapse
|
28
|
Kokkorakis M, Muzurović E, Volčanšek Š, Chakhtoura M, Hill MA, Mikhailidis DP, Mantzoros CS. Steatotic Liver Disease: Pathophysiology and Emerging Pharmacotherapies. Pharmacol Rev 2024; 76:454-499. [PMID: 38697855 DOI: 10.1124/pharmrev.123.001087] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 05/05/2024] Open
Abstract
Steatotic liver disease (SLD) displays a dynamic and complex disease phenotype. Consequently, the metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) therapeutic pipeline is expanding rapidly and in multiple directions. In parallel, noninvasive tools for diagnosing and monitoring responses to therapeutic interventions are being studied, and clinically feasible findings are being explored as primary outcomes in interventional trials. The realization that distinct subgroups exist under the umbrella of SLD should guide more precise and personalized treatment recommendations and facilitate advancements in pharmacotherapeutics. This review summarizes recent updates of pathophysiology-based nomenclature and outlines both effective pharmacotherapeutics and those in the pipeline for MASLD/MASH, detailing their mode of action and the current status of phase 2 and 3 clinical trials. Of the extensive arsenal of pharmacotherapeutics in the MASLD/MASH pipeline, several have been rejected, whereas other, mainly monotherapy options, have shown only marginal benefits and are now being tested as part of combination therapies, yet others are still in development as monotherapies. Although the Food and Drug Administration (FDA) has recently approved resmetirom, additional therapeutic approaches in development will ideally target MASH and fibrosis while improving cardiometabolic risk factors. Due to the urgent need for the development of novel therapeutic strategies and the potential availability of safety and tolerability data, repurposing existing and approved drugs is an appealing option. Finally, it is essential to highlight that SLD and, by extension, MASLD should be recognized and approached as a systemic disease affecting multiple organs, with the vigorous implementation of interdisciplinary and coordinated action plans. SIGNIFICANCE STATEMENT: Steatotic liver disease (SLD), including metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis, is the most prevalent chronic liver condition, affecting more than one-fourth of the global population. This review aims to provide the most recent information regarding SLD pathophysiology, diagnosis, and management according to the latest advancements in the guidelines and clinical trials. Collectively, it is hoped that the information provided furthers the understanding of the current state of SLD with direct clinical implications and stimulates research initiatives.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Emir Muzurović
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Špela Volčanšek
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Marlene Chakhtoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Michael A Hill
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Dimitri P Mikhailidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| |
Collapse
|
29
|
Ossima AN, Brzustowski A, Paradis V, Van Beers B, Postic C, Laouénan C, Pol S, Castéra L, Gautier JF, Czernichow S, Vallet-Pichard A, Larger E, Serfaty L, Zins M, Valla D, Zaleski ID. Factors associated with high costs of patients with metabolic dysfunction-associated steatotic liver disease: an observational study using the French CONSTANCES cohort. Clin Diabetes Endocrinol 2024; 10:9. [PMID: 38659082 PMCID: PMC11044468 DOI: 10.1186/s40842-023-00163-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/07/2023] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND & AIMS Despite its high prevalence in the western world metabolic dysfunction-associated steatotic liver disease (MASLD) does not benefit from targeted pharmacological therapy. We measured healthcare utilisation and identified factors associated with high-cost MASLD patients in France. METHODS The prevalent population with MASLD (including non-alcoholic steatohepatitis) in the CONSTANCES cohort, a nationally representative sample of 200,000 adults aged between 18 and 69, was linked to the French centralised national claims database (SNDS). Study participants were identified by the fatty liver index (FLI) over the period 2015-2019. MASLD individuals were classified according as "high-cost" (above 90th percentile) or "non-high cost" (below 90th percentile). Factors significantly associated with high costs were identified using a multivariate logistic regression model. RESULTS A total of 14,437 predominantly male (69%) participants with an average age of 53 ± SD 12 years were included. They mainly belonged to socially deprived population groups with co-morbidities such as diabetes, high blood pressure, mental health disorders and cardiovascular complications. The average expenditure was €1860 ± SD 4634 per year. High-cost MASLD cost €10,863 ± SD 10,859 per year. Conditions associated with high-cost were mental health disorders OR 1.79 (1.44-2.22), cardiovascular diseases OR 1.54 (1.21-1.95), metabolic comorbidities OR 1.50 (1.25-1.81), and respiratory disease OR 1.50 (1.11-2.00). The 10% high-cost participants accounted for 58% of the total national health care expenditures for MASLD. CONCLUSION Our results emphasize the need for comprehensive management of the comorbid conditions which were the major cost drivers of MASLD.
Collapse
Affiliation(s)
- Arnaud Nze Ossima
- DRCI- Health economics, Assistance Publique-Hôpitaux de Paris, Hôpital de l'Hôtel Dieu, 75004, Paris, France
| | - Angélique Brzustowski
- Université Paris Cité, INSERM, Centre de recherche sur l'inflammation, F-75018, Paris, France
| | - Valérie Paradis
- Université Paris Cité, Paris, France AP-HP, Hôpital Beaujon, 92110 Clichy, France Service Anatomie et cytologie pathologiques, INSERM, Centre de recherche sur l'inflammation, F-75018, Paris, France
| | | | - Catherine Postic
- Université Paris Cité, INSERM, Centre de recherche sur l'inflammation, F-75018, Paris, France
| | - Cédric Laouénan
- Université Paris Cité, INSERM, IAME UMR 1137, Paris, France, AP-HP.Nord, Hôpital Bichat, Département d'Epidémiologie Biostatistique et Recherche Clinique, Paris, France
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France AP-HP, Hôpital Bichat Service DEBRC, 75018, Paris, France
| | - Stanislas Pol
- Liver department, Hôpital Cochin-APHP, Université Paris Cité, Paris, France
| | - Laurent Castéra
- Hepatology department, Hôpital Beaujon, AP-HP, Université Paris Cité, INSERM UMR 1149, CRI, Clichy, France
| | - Jean-François Gautier
- Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière group and Inserm U1151, Service de diabétologie et d'endocrinologie - Centre Universitaire du Diabète et de ses Complications, Paris, France
| | - Sebastien Czernichow
- Université de Paris-Cité and Université Sorbonne Paris Nord, Paris, France, Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Nutrition, Centre Spécialisé Obésité, Hôpital Européen Georges Pompidou, Paris, France, Centre of Research in Epidemiology and Statistics (CRESS-U1153), Inserm, INRAE, Paris, France
| | | | - Etienne Larger
- Université Paris Cité, Diabetology department, Hôpital Cochin-APHP, Paris, France
| | - Lawrence Serfaty
- Université de Strasbourg, Hepatogastroenterology Service, Hôpital Hautepierre, Hôpitaux Universitaires de Strasbourg 67000, Strasbourg, France, INSERM UMR_S938, Sorbonne Université, Paris, France
| | - Marie Zins
- UMS 11 Inserm, Versailles-Saint Quentin University, Versailles, France
| | - Dominique Valla
- Université Paris Cité, INSERM, Centre de recherche sur l'inflammation, F-75018, Paris, France
- Service hépatologie, AP-HP, Hôpital Beaujon, 92110, Clichy, France
| | - Isabelle Durand Zaleski
- DRCI- Health economics, Assistance Publique-Hôpitaux de Paris, Hôpital de l'Hôtel Dieu, 75004, Paris, France.
- Universite Paris Est Créteil, Assistance Publique-Hôpitaux de Paris, Service de Santé Publique, Henri Mondor-Albert- Chenevier, 94000 Créteil, France, Centre of Research in Epidemiology and Statistics (CRESS-U1153),Inserm, INRAE, Paris, France.
| |
Collapse
|
30
|
Liu F, Bi M, Jing X, Ding H, Zeng J, Zheng R, Chen Y, Wang W, Xie X, Mi C, Chen M, Cheng W, Zhang S, Wang Z, Zhang C, Zhou H, Cheng Z, Han Z, Yu J, Liang P. Multiparametric US for Identifying Metabolic Dysfunction-associated Steatohepatitis: A Prospective Multicenter Study. Radiology 2024; 310:e232416. [PMID: 38501954 DOI: 10.1148/radiol.232416] [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: 03/20/2024]
Abstract
Background Noninvasive evaluation of metabolic dysfunction-associated fatty liver disease (MAFLD) with multiparametric US is essential, but multicenter studies are lacking. Purpose To evaluate the ability of multiparametric US with attenuation imaging (ATI) and two-dimensional (2D) shear-wave elastography (SWE) for predicting metabolic dysfunction-associated steatohepatitis (MASH) in participants with MAFLD, regardless of hepatitis B virus infection status. Materials and Methods This prospective cross-sectional multicenter study of consecutive adults with MAFLD who underwent multiparametric US with ATI and 2D SWE, as well as liver biopsy, from September 2020 to June 2022 was conducted in 12 tertiary hospitals in China. Multivariable logistic regression was performed to assess risk factors associated with MASH. Area under the receiver operating characteristic curve (AUC) analysis was used to evaluate diagnostic performance in predicting MASH in training and validation groups (6:4 ratio of participants), and for a post hoc subgroup analysis of hepatitis B virus infection and diabetes. Results A total of 424 participants (median age, 47 years; IQR, 34-59 years; 244 male) were evaluated, including 332 participants (78%) with MASH and 92 (22%) without. Attenuation coefficient (AC) (odds ratio [OR], 3.32 [95% CI: 1.94, 5.71]; P < .001), alanine aminotransferase (ALT) level (OR, 4.42 [95% CI: 1.78, 10.94]; P = .001), and international normalized ratio (INR) (OR, 0.59 [95% CI: 0.37, 0.95]; P = .03) were independently associated with MASH. A combined model (AC, ALT, and INR) had AUCs of 0.85 (95% CI: 0.79, 0.91) and 0.77 (95% CI: 0.69, 0.85) for predicting MASH in the training and validation groups, respectively. AUC values for the subgroups with and without diabetes were 0.83 (95% CI: 0.72, 0.94) and 0.81 (95% CI: 0.75, 0.87) and for the subgroups with and without hepatitis B were 0.82 (95% CI: 0.74, 0.90) and 0.79 (95% CI: 0.71, 0.87), respectively. Conclusion A model combining AC, ALT level, and INR showed good discrimination ability for predicting MASH in participants with MAFLD. Clinical trial registration no. NCT04551716 © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Reuter in this issue.
Collapse
Affiliation(s)
- Fangyi Liu
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Mingsen Bi
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Xiang Jing
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Hong Ding
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Jie Zeng
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Rongqin Zheng
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Yaqing Chen
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Wenping Wang
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Xiaoyan Xie
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Chengrong Mi
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Man Chen
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Wen Cheng
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Shuhua Zhang
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Zhanbo Wang
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Chunquan Zhang
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Hongyu Zhou
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Zhigang Cheng
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Zhiyu Han
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Jie Yu
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| | - Ping Liang
- From the Department of Interventional Ultrasound, Fifth Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Rd, Beijing 100853, China (F.L., M.B., Z.C., Z.H., J.Y., P.L.); Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China (X.J., H.Z.); Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China (H.D.); Department of Medical Ultrasound, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.Z., R.Z.); Department of Ultrasound in Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China (Y.C.); Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China (W.W.); Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (X.X.); Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, China (C.M.); Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.C.); Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China (W.C.); Department of Ultrasound, North China University of Science and Technology Affiliated Hospital, Tangshan, China (S.Z.); Department of Pathology, First Medical Center, Chinese PLA General Hospital, Beijing, China (Z.W.); and Department of Ultrasound, The Second Affiliated Hospital of Nanchang University, Nanchang, China (C.Z.)
| |
Collapse
|
31
|
Cusi K. Nonalcoholic Fatty Liver Disease in Diabetes: A Call to Action. Diabetes Spectr 2024; 37:5-7. [PMID: 38385097 PMCID: PMC10877210 DOI: 10.2337/dsi23-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Affiliation(s)
- Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL
| |
Collapse
|
32
|
Cusi K, Budd J, Johnson E, Shubrook J. Making Sense of the Nonalcoholic Fatty Liver Disease Clinical Practice Guidelines: What Clinicians Need to Know. Diabetes Spectr 2024; 37:29-38. [PMID: 38385100 PMCID: PMC10877212 DOI: 10.2337/dsi23-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Standards of care summarized in clinical practice guidelines for nonalcoholic fatty liver disease (NAFLD) offer clinicians a streamlined diagnostic and management approach based on the best available evidence. These recommendations have changed a great deal in recent years; today, there is a clear focus on screening for the early identification and risk stratification of patients at high risk of steatohepatitis and clinically significant fibrosis to promote timely referrals to specialty care when needed. This article reviews and provides the rationale for current guidelines for NAFLD screening, diagnosis, treatment, and monitoring and addresses barriers to providing evidence-based NAFLD care and how to overcome them. The current paradigm of care calls for primary care clinicians and specialists to work together, within a multidisciplinary care team familiar with obesity and diabetes care, to provide comprehensive management of these complex patients.
Collapse
Affiliation(s)
- Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL
| | - Jeff Budd
- Division of General Internal Medicine, University of Florida, Gainesville, FL
| | - Eric Johnson
- Department of Family and Community Medicine, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND
| | - Jay Shubrook
- Department of Clinical Sciences and Community Health, Touro University California College of Osteopathic Medicine, Vallejo, CA
| |
Collapse
|
33
|
Nogueira JP, Cusi K. Role of Insulin Resistance in the Development of Nonalcoholic Fatty Liver Disease in People With Type 2 Diabetes: From Bench to Patient Care. Diabetes Spectr 2024; 37:20-28. [PMID: 38385099 PMCID: PMC10877218 DOI: 10.2337/dsi23-0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Insulin resistance is implicated in both the pathogenesis of nonalcoholic fatty liver disease (NAFLD) and its progression from steatosis to steatohepatitis, cirrhosis, and even hepatocellular carcinoma, which is known to be more common in people with type 2 diabetes. This article reviews the role of insulin resistance in the metabolic dysfunction observed in obesity, type 2 diabetes, atherogenic dyslipidemia, and hypertension and how it is a driver of the natural history of NAFLD by promoting glucotoxicity and lipotoxicity. The authors also review the genetic and environmental factors that stimulate steatohepatitis and fibrosis progression and their relationship with cardiovascular disease and summarize guidelines supporting the treatment of NAFLD with diabetes medications that reduce insulin resistance, such as pioglitazone or glucagon-like peptide 1 receptor agonists.
Collapse
Affiliation(s)
- Juan Patricio Nogueira
- Universidad del Pacifico, Asunción, Paraguay
- Centro de Investigación en Endocrinología, Nutrición y Metabolismo, Facultad de Ciencias de la Salud, Universidad Nacional de Formosa, Formosa, Argentina
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL
| |
Collapse
|
34
|
Denimal D, Ponnaiah M, Jeannin AC, Phan F, Hartemann A, Boussouar S, Charpentier E, Redheuil A, Foufelle F, Bourron O. Non-alcoholic fatty liver disease biomarkers estimate cardiovascular risk based on coronary artery calcium score in type 2 diabetes: a cross-sectional study with two independent cohorts. Cardiovasc Diabetol 2024; 23:69. [PMID: 38351039 PMCID: PMC10865592 DOI: 10.1186/s12933-024-02161-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Studies have demonstrated that coronary artery calcification on one hand and non-alcoholic fatty liver disease (NAFLD) on the other hand are strongly associated with cardiovascular events. However, it remains unclear whether NAFLD biomarkers could help estimate cardiovascular risk in individuals with type 2 diabetes (T2D). The primary objective of the present study was to investigate whether the biomarkers of NAFLD included in the FibroMax® panels are associated with the degree of coronary artery calcification in patients with T2D. METHODS A total of 157 and 460 patients with T2D were included from the DIACART and ACCoDiab cohorts, respectively. The coronary artery calcium score (CACS) was measured in both cohorts using computed tomography. FibroMax® panels (i.e., SteatoTest®, FibroTest®, NashTest®, and ActiTest®) were determined from blood samples as scores and stages in the DIACART cohort and as stages in the ACCoDiab cohort. RESULTS CACS significantly increased with the FibroTest® stages in both the DIACART and ACCoDiab cohorts (p-value for trend = 0.0009 and 0.0001, respectively). In DIACART, the FibroTest® score was positively correlated with CACS in univariate analysis (r = 0.293, p = 0.0002) and remained associated with CACS independently of the traditional cardiovascular risk factors included in the SCORE2-Diabetes model [β = 941 ± 425 (estimate ± standard error), p = 0.028]. In the ACCoDiab cohort, the FibroTest® F3-F4 stage was positively correlated with CACS in point-biserial analysis (rpbi = 0.104, p = 0.024) and remained associated with CACS after adjustment for the traditional cardiovascular risk factors included in the SCORE2-Diabetes model (β = 234 ± 97, p = 0.016). Finally, the prediction of CACS was improved by adding FibroTest® to the traditional cardiovascular risk factors included in the SCORE2-Diabetes model (goodness-of-fit of prediction models multiplied by 4.1 and 6.7 in the DIACART and ACCoDiab cohorts, respectively). In contrast, no significant relationship was found between FibroMax® panels other than FibroTest® and CACS in either cohort. CONCLUSIONS FibroTest® is independently and positively associated with the degree of coronary artery calcification in patients with T2D, suggesting that FibroTest® could be a relevant biomarker of coronary calcification and cardiovascular risk. TRIAL REGISTRATION ClinicalTrials.gov identifiers NCT02431234 and NCT03920683.
Collapse
Affiliation(s)
- Damien Denimal
- Center for Translational and Molecular Medicine, INSERM UMR 1231, Dijon, France
- Department of Clinical Biochemistry, Dijon Bourgogne University Hospital, Dijon, France
| | | | - Anne-Caroline Jeannin
- Sorbonne Université, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
| | - Franck Phan
- Sorbonne Université, Paris, France
- Centre de Recherche des Cordeliers, INSERM UMR_S 1138, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
| | - Agnès Hartemann
- Centre de Recherche des Cordeliers, INSERM UMR_S 1138, Paris, France
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France
| | - Samia Boussouar
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Laboratoire d'Imagerie Biomédicale INSERM_1146, CNRS_7371, Paris, France
- ICT Cardiovascular and Thoracic Imaging Unit, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié Salpêtrière University Hospital, Paris, France
| | - Etienne Charpentier
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Laboratoire d'Imagerie Biomédicale INSERM_1146, CNRS_7371, Paris, France
- ICT Cardiovascular and Thoracic Imaging Unit, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié Salpêtrière University Hospital, Paris, France
| | - Alban Redheuil
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- Laboratoire d'Imagerie Biomédicale INSERM_1146, CNRS_7371, Paris, France
- ICT Cardiovascular and Thoracic Imaging Unit, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié Salpêtrière University Hospital, Paris, France
| | - Fabienne Foufelle
- Centre de Recherche des Cordeliers, INSERM UMR_S 1138, Paris, France
| | - Olivier Bourron
- Sorbonne Université, Paris, France.
- Centre de Recherche des Cordeliers, INSERM UMR_S 1138, Paris, France.
- Department of Diabetology, Assistance Publique‑Hôpitaux de Paris (APHP), Pitié-Salpêtrière Hospital, 47‑83 Boulevard de l'Hôpital, Paris, France.
| |
Collapse
|
35
|
Bech KT, Lindvig KP, Thiele M, Castera L. Algorithms for Early Detection of Silent Liver Fibrosis in the Primary Care Setting. Semin Liver Dis 2024; 44:23-34. [PMID: 38262447 DOI: 10.1055/s-0043-1778127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
More than one-third of the adult world population has steatotic liver disease (SLD), with a few percent of individuals developing cirrhosis after decades of silent liver fibrosis accumulation. Lack of systematic early detection causes most patients to be diagnosed late, after decompensation, when treatment has limited effect and survival is poor. Unfortunately, no isolated screening test in primary care can sufficiently predict advanced fibrosis from SLD. Recent efforts, therefore, combine several parameters into screening algorithms, to increase diagnostic accuracy. Besides patient selection, for example, by specific characteristics, algorithms include nonpatented or patented blood tests and liver stiffness measurements using elastography-based techniques. Algorithms can be composed as a set of sequential tests, as recommended by most guidelines on primary care pathways. Future use of algorithms that are easy to interpret, cheap, and semiautomatic will improve the management of patients with SLD, to the benefit of global health care systems.
Collapse
Affiliation(s)
- Katrine Tholstrup Bech
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Katrine Prier Lindvig
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Maja Thiele
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Laurent Castera
- Service d'Hépatologie, Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Beaujon, Clichy, France
- Faculté de Médecine, Université Paris Cité, UMR1149 (CRI), INSERM, Paris, France
| |
Collapse
|
36
|
Xu J, Xia Q, Wu T, Shao Y, Wang Y, Jin N, Tian P, Wu L, Lu X. Prophylactic treatment with Bacteroides uniformis and Bifidobacterium bifidum counteracts hepatic NK cell immune tolerance in nonalcoholic steatohepatitis induced by high fat diet. Gut Microbes 2024; 16:2302065. [PMID: 38196273 PMCID: PMC10793665 DOI: 10.1080/19490976.2024.2302065] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Hepatic immunity is one of the driving forces for the development of nonalcoholic steatohepatitis (NASH), and targeting gut microbiota is believed to affect the hepatic immune constitution. Here, we aimed to investigate the hepatic immunological state in NASH, with a specific emphasis on natural killer (NK) cells. In addition, we aimed to identify the contributing species that target hepatic immunity to provide new directions and support the feasibility of immunotherapy for NASH. A possible NASH population was determined by combination of long-term severe fatty liver, metabolic disorders and increased serum CK18 to detect serum immune factors and gut microbiota. NASH was induced in mice fed a high-fat diet to verify the prophylactic effect of the functional species on the immunopathology and development of NASH. Hepatic immunologic state was examined, and the effector functions of NK cells were detected. Hepatic transcriptome, proteomic, and fecal metagenome were performed. We observed a statistical increase in serum IL-10 (p < 0.001) and non-statistical decrease in interferon-γ and IL-6 in NASH population, hinting at the possibility of immune tolerance. Fecal Bacteroides uniformis and Bifidobacterium bifidum were abundant in healthy population but depleted in NASH patients. In NASH mice, hepatic CD8+T cells, macrophages, and dendritic cells were increased (p < 0.01), and NK cells were inhibited, which were identified with decreased granzyme B (p < 0.05). Bacteroides uniformis and Bifidobacterium bifidum improved hepatic pathological and metabolic cues, increased hepatic NK cells and reduced macrophages (p < 0.05). Bacteroides uniformis also restored hepatic NK cell function, which was identified as increased CD107a (p < 0.05). Transcriptional and translational profiling revealed that the functional species might restore the function of hepatic NK cells through multiple pathways, such as reduction of inhibitory molecules in NK cells. Bacteroides uniformis and Bifidobacterium bifidum are novel prophylactics for NASH that restore the impaired function of hepatic NK cells.
Collapse
Affiliation(s)
- Jingyuan Xu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qiaoyun Xia
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Ting Wu
- Department of Citizen Health, Community Health Service Center of Jinxi Town, Kunshan, China
| | - Yong Shao
- Department of Citizen Health, Community Health Service Center of Jinxi Town, Kunshan, China
| | - Yatao Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Nuyun Jin
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Peiying Tian
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Longyun Wu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaolan Lu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
37
|
Abeysekera KWM, Valenti L, Younossi Z, Dillon JF, Allen AM, Nourredin M, Rinella ME, Tacke F, Francque S, Ginès P, Thiele M, Newsome PN, Guha IN, Eslam M, Schattenberg JM, Alqahtani SA, Arrese M, Berzigotti A, Holleboom AG, Caussy C, Cusi K, Roden M, Hagström H, Wong VWS, Mallet V, Castera L, Lazarus JV, Tsochatzis EA. Implementation of a liver health check in people with type 2 diabetes. Lancet Gastroenterol Hepatol 2024; 9:83-91. [PMID: 38070521 DOI: 10.1016/s2468-1253(23)00270-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 12/18/2023]
Abstract
As morbidity and mortality related to potentially preventable liver diseases are on the rise globally, early detection of liver fibrosis offers a window of opportunity to prevent disease progression. Early detection of non-alcoholic fatty liver disease allows for initiation and reinforcement of guidance on bodyweight management, risk stratification for advanced liver fibrosis, and treatment optimisation of diabetes and other metabolic complications. Identification of alcohol-related liver disease provides the opportunity to support patients with detoxification and abstinence programmes. In all patient groups, identification of cirrhosis ensures that patients are enrolled in surveillance programmes for hepatocellular carcinoma and portal hypertension. When considering early detection strategies, success can be achieved from applying ad-hoc screening for liver fibrosis in established frameworks of care. Patients with type 2 diabetes are an important group to consider case findings of advanced liver fibrosis and cirrhosis, as up to 19% have advanced fibrosis (which is ten times higher than the general population) and almost 70% have non-alcoholic fatty liver disease. Additionally, patients with type 2 diabetes with alcohol use disorders have the highest proportion of liver-related morbidity of people with type 2 diabetes generally. Patients with type 2 diabetes receive an annual diabetes review as part of their routine clinical care, in which the health of many organs are considered. Yet, liver health is seldom included in this review. This Viewpoint argues that augmenting the existing risk stratification strategy with an additional liver health check provides the opportunity to detect advanced liver fibrosis, thereby opening a window for early interventions to prevent end-stage liver disease and its complications, including hepatocellular carcinoma.
Collapse
Affiliation(s)
- Kushala W M Abeysekera
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, UK; Department of Liver Medicine, Bristol Royal Infirmary, Bristol, UK
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy; Precision Medicine, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Zobair Younossi
- Beatty Liver and Obesity Research Program, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
| | - John F Dillon
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Alina M Allen
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Mazen Nourredin
- Sherrie & Alan Conover Center for Liver Disease & Transplantation, Underwood Center for Digestive Disorders, Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA; Houston Research Institute, Houston, Texas, USA
| | - Mary E Rinella
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Edegem, Belgium; Translational Sciences in Inflammation and Immunology, Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Care Sciences, University of Antwerp, Antwerp, Belgium
| | - Pere Ginès
- Liver Unit, Hospital Clinic Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, Spain; Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Maja Thiele
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Department for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Philip N Newsome
- National Institute for Health Research, Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Indra Neil Guha
- National Institute for Health Research, Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, Department of Medicine, University Medical Center, Mainz, Germany
| | - Saleh A Alqahtani
- Liver Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia; Division of Gastroenterology & Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adriaan G Holleboom
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Cyrielle Caussy
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, University of Lyon, Lyon, France; Department of Endocrinology, Diabetes and Nutrition, Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Munich, Germany
| | - Hannes Hagström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Vincent Mallet
- Faculty of Medicine, Université Paris Cité, F-75006, Paris, France; Service d'Hépatologie, Département Médico-Universitaire Cancérologie et Spécialités Médico-Chirurgicales, AP-HP.Centre, Groupe Hospitalier Cochin Port Royal, Paris, France
| | - Laurent Castera
- Department of Hepatology, Beaujon Hospital, Université Paris Cité, INSERM UMR1149, Paris, France
| | - Jeffrey V Lazarus
- Department of Health Policy and Mangement, City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA; Barcelona Institute for Global Health, Hospital Clinic Barcelona, University of Barcelona, Barcelona, Spain
| | - Emmanuel A Tsochatzis
- Sheila Sherlock Liver Unit, Royal Free Hospital, London, UK; UCL Institute of Liver and Digestive Health, University College London, UK.
| |
Collapse
|
38
|
Moein HR, Mohammed S. Not All "Normal" Liver Enzymes Are Normal! Gastroenterology 2023; 165:1586-1587. [PMID: 37716375 DOI: 10.1053/j.gastro.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Affiliation(s)
- Hamid Reza Moein
- Department of Gastroenterology, UNC Health Blue Ridge, Morganton, North Carolina
| | - Suneel Mohammed
- Department of Gastroenterology, UNC Health Blue Ridge, Morganton, North Carolina
| |
Collapse
|
39
|
Poynard T, Deckmyn O, Peta V, Paradis V, Gautier JF, Brzustowski A, Bedossa P, Castera L, Pol S, Valla D. Prospective direct comparison of non-invasive liver tests in outpatients with type 2 diabetes using intention-to-diagnose analysis. Aliment Pharmacol Ther 2023; 58:888-902. [PMID: 37642160 DOI: 10.1111/apt.17688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/23/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND No prospective diagnostic studies have directly compared widespread non-invasive liver tests in patients with type 2 diabetes (T2D) using the intention-to-diagnose method for each of the three main histological features of metabolic dysfunction associated steatotic liver disease - namely fibrosis, metabolic dysfunction-associated steatohepatitis (MASH), and steatosis. AIMS To compare the performance of nine tests using the intention-to-diagnose rather than the standard method, which would exclude non-evaluable participants METHODS: Biopsy was used as the reference with predetermined cut-offs, advanced fibrosis being the main endpoint. The Nash-FibroTest panel including FibroTest-T2D, SteatoTest-T2D and MashTest-T2D was optimised for type 2 diabetes. FibroTest-T2D was compared to vibration-controlled transient elastography stiffness (VCTE), two-dimensional shear-wave elastography stiffness (TD-SWE), and Fibrosis-4 blood test. NashTest-T2D was compared to aspartate aminotransferase. SteatoTest-T2D was compared to the controlled attenuation parameter and the hepatorenal gradient. RESULTS Among 402 cases, non-evaluable tests were 6.7% for VCTE, 4.0% for hepatorenal gradient, 3.2% for controlled attenuation parameter, 1.5% for TD-SWE, 1.2% for NashTest-T2D, and 0.02% for Fibrosis-4, aspartate aminotransferase and SteatoTest-T2D. The VCTE AUROC for advanced fibrosis was over-estimated by 6% (0.83 [95% CI: 0.78-0.87]) by standard analysis compared to intention-to-diagnose (0.77 [0.72-0.81] p = 0.008). The AUROCs for advanced fibrosis did not differ significantly in intention-to-diagnose between FibroTest-T2D (0.77; 95% CI: 0.73-0.82), VCTE (0.77; 95% CI: 0.72-0.81) and TD-SWE(0.78; 0.74-0.83) but were all higher than the Fibrosis-4 score (0.70; 95% CI all differences ≥7%; p ≤ 0.03). For MASH, MashTest-T2D had a higher AUROC (0.76; 95% CI: 0.70-0.80) than aspartate aminotransferase (0.72; 95% CI: 0.66-0.77; p = 0.035). For steatosis, AUROCs did not differ significantly between SteatoTest-T2D, controlled attenuation parameter and hepatorenal gradient. CONCLUSIONS In intention-to-diagnose analysis, FibroTest-T2D, TD-SWE and VCTE performed similarly for staging fibrosis, and out-performed Fibrosis-4 in outpatients with type 2 diabetes. The standard analysis over-estimated VCTE performance. CLINICALTRIAL gov: NCT03634098.
Collapse
Affiliation(s)
- Thierry Poynard
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Paris, France
- BioPredictive, Paris, France
| | | | | | - Valérie Paradis
- Department of Pathology, AP-HP, Beaujon Hospital, Clichy, France
| | - Jean-Francois Gautier
- Department of Diabetes and Endocrinology, APHP, INSERM U1138, Hôpital Lariboisière, Paris, France
| | | | - Pierre Bedossa
- Department of Pathology, AP-HP, Beaujon Hospital, Clichy, France
| | - Laurent Castera
- Department of Hepatology, AP-HP, Beaujon Hospital, Clichy, France
| | - Stanislas Pol
- Department of Hepatology, Cochin Hospital, Université Paris Descartes, Paris, France
| | - Dominique Valla
- Department of Hepatology, AP-HP, Beaujon Hospital, Clichy, France
| |
Collapse
|
40
|
Romero-Gómez M, Zelber-Sagi S, Martín F, Bugianesi E, Soria B. Nutrition could prevent or promote non-alcoholic fatty liver disease: an opportunity for intervention. BMJ 2023; 383:e075179. [PMID: 37813416 PMCID: PMC10561058 DOI: 10.1136/bmj-2023-075179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Affiliation(s)
- Manuel Romero-Gómez
- UCM Digestive Diseases and Ciberehd. Virgen del Rocío University Hospital, Spain
- Institute of Biomedicine of Seville (HUVR/CSIC/US), Spain
- Department of Medicine, University of Seville, Seville, Spain
| | - Shira Zelber-Sagi
- School of Public Health, University of Haifa, Haifa, Israel
- Department of Gastroenterology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Franz Martín
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Pablo de Olavide University, University of Seville, Spain
- Biomedical Research Network on Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, Turin, Italy
- Division of Gastroenterology, Città Della Salute e Della Scienza University-Hospital, Turin, Italy
| | - Bernat Soria
- Unit of Advanced Therapies and Regenerative Medicine, Health Research Institute of the Dr Balmis University Hospital of Alicante (ISABIAL), Institute of Bioengineering University Miguel Hernández, CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Hammoutene A, Laouirem S, Albuquerque M, Colnot N, Brzustowski A, Valla D, Provost N, Delerive P, Paradis V. A new NRF2 activator for the treatment of human metabolic dysfunction-associated fatty liver disease. JHEP Rep 2023; 5:100845. [PMID: 37663119 PMCID: PMC10472315 DOI: 10.1016/j.jhepr.2023.100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/09/2023] [Accepted: 06/20/2023] [Indexed: 09/05/2023] Open
Abstract
Background & Aims Oxidative stress triggers metabolic-associated fatty liver disease (MAFLD) and fibrosis. Previous animal studies demonstrated that the transcription factor nuclear factor (erythroid-derived 2)-like 2 (NRF2), the master regulator of antioxidant response, protects against MAFLD and fibrosis. S217879, a next generation NRF2 activator has been recently shown to trigger diet-induced steatohepatitis resolution and to reduce established fibrosis in rodents. Our aim was to evaluate the therapeutic potential of S217879 in human MAFLD and its underlying mechanisms using the relevant experimental 3D model of patient-derived precision cut liver slices (PCLS). Methods We treated PCLS from 12 patients with varying stages of MAFLD with S217879 or elafibranor (peroxisome proliferator-activated receptor [PPAR]α/δ agonist used as a referent molecule) for 2 days. Safety and efficacy profiles, steatosis, liver injury, inflammation, and fibrosis were assessed as well as mechanisms involved in MAFLD pathophysiology, namely antioxidant response, autophagy, and endoplasmic reticulum-stress. Results Neither elafibranor nor S217879 had toxic effects at the tested concentrations on human PCLS with MAFLD. PPARα/δ and NRF2 target genes (pyruvate dehydrogenase kinase 4 [PDK4], fibroblast growth factor 21 [FGF21], and NAD(P)H quinone dehydrogenase 1 [NQO1], heme oxygenase 1 [HMOX1], respectively) were strongly upregulated in PCLS in response to elafibranor and S217879, respectively. Compared with untreated PCLS, elafibranor and S217879-treated slices displayed lower triglycerides and reduced inflammation (IL-1β, IL-6, chemokine (C-C motif) ligand 2 [CCL2]). Additional inflammatory markers (chemokine (C-C motif) ligand 5 [CCL5], stimulator of interferon genes [STING], intercellular adhesion molecule-1 [ICAM-1], vascular cell adhesion molecule-1 [VCAM-1]) were downregulated by S217879. S217879 but not elafibranor lowered DNA damage (phospho-Histone H2A.X [p-H2A.X], RAD51, X-ray repair cross complementing 1 [XRCC1]) and apoptosis (cleaved caspase-3), and inhibited fibrogenesis markers expression (alpha smooth muscle actin [α-SMA], collagen 1 alpha 1 [COL1A1], collagen 1 alpha 2 [COL1A2]). Such effects were mediated through an improvement of lipid metabolism, activated antioxidant response and enhanced autophagy, without effect on endoplasmic reticulum-stress. Conclusions This study highlights the therapeutic potential of a new NRF2 activator for MAFLD using patient-derived PCLS supporting the evaluation of NRF2 activating strategies in clinical trials. Impact and implications Oxidative stress is a major driver of metabolic-associated fatty liver disease (MAFLD) development and progression. Nuclear factor (erythroid-derived 2)-like 2, the master regulator of the antioxidative stress response, is an attractive therapeutic target for the treatment of MAFLD. This study demonstrates that S217879, a new potent and selective nuclear factor (erythroid-derived 2)-like 2 activator, displays antisteatotic effects, lowers DNA damage, apoptosis, and inflammation and inhibits fibrogenesis in human PCLS in patients with MAFLD.
Collapse
Affiliation(s)
- Adel Hammoutene
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
- Cardiovascular and Metabolic Diseases Research, Institut de Recherches Servier, Suresnes, France
| | - Samira Laouirem
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
| | - Miguel Albuquerque
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
- Département de Pathologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Nathalie Colnot
- Département de Pathologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Angélique Brzustowski
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
| | - Dominique Valla
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
| | - Nicolas Provost
- Cardiovascular and Metabolic Diseases Research, Institut de Recherches Servier, Suresnes, France
| | - Philippe Delerive
- Cardiovascular and Metabolic Diseases Research, Institut de Recherches Servier, Suresnes, France
| | - Valérie Paradis
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
- Département de Pathologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - the QUID-NASH Research Group
- Université Paris Cité, Inserm, Centre de Recherche sur l'inflammation, F-75018, Paris, France
- Cardiovascular and Metabolic Diseases Research, Institut de Recherches Servier, Suresnes, France
- Département de Pathologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| |
Collapse
|
42
|
Ciardullo S, Vergani M, Perseghin G. Nonalcoholic Fatty Liver Disease in Patients with Type 2 Diabetes: Screening, Diagnosis, and Treatment. J Clin Med 2023; 12:5597. [PMID: 37685664 PMCID: PMC10488336 DOI: 10.3390/jcm12175597] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently renamed metabolic dysfunction-associated steatotic liver disease (MASLD) affects ~70% of patients with type 2 diabetes (T2D), with ~20% showing signs of advanced liver fibrosis. Patients with T2D are at an increased risk of developing cirrhosis, liver failure, and hepatocellular carcinoma and their liver-related mortality is doubled compared with non-diabetic individuals. Nonetheless, the condition is frequently overlooked and disease awareness is limited both among patients and among physicians. Given recent epidemiological evidence, clinical practice guidelines recommend screening for NAFLD/MASLD and advanced liver fibrosis in patients with T2D. While many drugs are currently being tested for the treatment of NAFLD/MASLD, none of them have yet received formal approval from regulatory agencies. However, several classes of antidiabetic drugs (namely pioglitazone, sodium-glucose transporter 2 inhibitors, glucagon-like peptide 1 receptor agonists, and multi-agonists) have shown favorable effects in terms of liver enzymes, liver fat content and, in some occasions, on histologic features such as inflammation and fibrosis. Therefore, diabetologists have the opportunity to actively treat NAFLD/MASLD, with a concrete possibility of changing the natural history of the disease. In the present narrative review, we summarize evidence and clinical recommendations for NAFLD/MAFLD screening in the setting of T2D, as well as on the effect of currently available glucose-lowering drugs on hepatic endpoints.
Collapse
Affiliation(s)
- Stefano Ciardullo
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900 Monza, MB, Italy; (M.V.); (G.P.)
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, MI, Italy
| | - Michela Vergani
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900 Monza, MB, Italy; (M.V.); (G.P.)
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, MI, Italy
| | - Gianluca Perseghin
- Department of Medicine and Rehabilitation, Policlinico di Monza, Via Modigliani 10, 20900 Monza, MB, Italy; (M.V.); (G.P.)
- Department of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, MI, Italy
| |
Collapse
|
43
|
Scoditti E, Marchesini G, Gastaldelli A. Hunting for Progressive NAFLD in Type 2 Diabetes: Do Not Trust Liver Enzymes! Diabetes Care 2023; 46:1332-1334. [PMID: 37339349 DOI: 10.2337/dci23-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Affiliation(s)
- Egeria Scoditti
- 1Institute of Clinical Physiology, National Research Council, Lecce, Italy
| | - Giulio Marchesini
- 2Department of Medical and Surgical Sciences, Alma Mater University, Bologna, Italy
- 3Azienda Ospedaliera di Bologna Sant'Orsola-Malpighi, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Bologna, Italy
| | - Amalia Gastaldelli
- 4Institute of Clinical Physiology, National Research Council, Pisa, Italy
| |
Collapse
|
44
|
Castera L, Cusi K. Diabetes and cirrhosis: Current concepts on diagnosis and management. Hepatology 2023; 77:2128-2146. [PMID: 36631005 DOI: 10.1097/hep.0000000000000263] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/03/2022] [Indexed: 01/13/2023]
Abstract
Type 2 diabetes mellitus is often associated with cirrhosis as comorbidities, acute illness, medications, and other conditions profoundly alter glucose metabolism. Both conditions are closely related in NAFLD, the leading cause of chronic liver disease, and given its rising burden worldwide, management of type 2 diabetes mellitus in cirrhosis will be an increasingly common dilemma. Having diabetes increases cirrhosis-related complications, including HCC as well as overall mortality. In the absence of effective treatments for cirrhosis, patients with type 2 diabetes mellitus should be systematically screened as early as possible for NAFLD-related fibrosis/cirrhosis using noninvasive tools, starting with a FIB-4 index followed by transient elastography, if available. In people with cirrhosis, an early diagnosis of diabetes is critical for an optimal management strategy (ie, nutritional goals, and glycemic targets). Diagnosis of diabetes may be missed if based on A1C in patients with cirrhosis and impaired liver function (Child-Pugh B-C) as anemia may turn the test unreliable. Clinicians must also become aware of their high risk of hypoglycemia, especially in decompensated cirrhosis where insulin is the only therapy. Care should be within multidisciplinary teams (nutritionists, obesity management teams, endocrinologists, hepatologists, and others) and take advantage of novel glucose-monitoring devices. Clinicians should become familiar with the safety and efficacy of diabetes medications for patients with advanced fibrosis and compensated cirrhosis. Management is conditioned by whether the patient has either compensated or decompensated cirrhosis. This review gives an update on the complex relationship between cirrhosis and type 2 diabetes mellitus, with a focus on its diagnosis and treatment, and highlights knowledge gaps and future directions.
Collapse
Affiliation(s)
- Laurent Castera
- Departement of Hepatology, Hospital Beaujon, Assistance Publique-Hôpitaux de Paris, INSERM UMR 1149, Université Paris Cité, Clichy, France
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, The University of Florida, Gainesville, Florida, USA
| |
Collapse
|