1
|
Jang SY, Yoon KT, Cho YY, Jo HG, Baek YH, Moon SY, Jo AJ, Kweon YO, Park SY, Lee YR, Jun DW, Tak WY. Aspartate aminotransferase-to-platelet ratio index outperforms Fibrosis-4 in 2843 Korean patients with metabolic dysfunction-associated steatotic liver disease. Hepatol Res 2025; 55:479-491. [PMID: 39612185 DOI: 10.1111/hepr.14143] [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: 09/20/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/30/2024]
Abstract
AIM The definition of metabolic dysfunction-associated steatotic liver disease (MASLD) has recently been proposed. We aim to investigate the diagnostic efficacy of noninvasive fibrosis markers in predicting liver fibrosis in patients with nonalcoholic fatty liver disease (NAFLD), metabolic dysfunction-associated fatty liver disease (MAFLD), and MASLD. METHODS This retrospective study involved 2843 patients diagnosed with steatotic liver disease at six tertiary hospitals in South Korea. Liver fibrosis was assessed using vibration-controlled transient elastography, and various noninvasive markers, including the aspartate aminotransferase-to-platelet ratio index (APRI), Fibrosis-4 index (FIB-4), NAFLD fibrosis score (NFS), and serum Mac-2-binding protein glycosylation isomer were analyzed. RESULTS Among 1106 patients, 79.9% met criteria for NAFLD, MAFLD, and MASLD. The APRI had area under the receiver operating characteristic curve (AUC) values of 0.819, 0.821, and 0.818 for liver fibrosis ≥F2, and 0.819, 0.824, and 0.884 for liver fibrosis ≥F3, and 0.890, 0.884, and 0.889 for fibrosis ≥F4 in NAFLD, MAFLD, and MASLD, respectively. The FIB-4 index showed AUC values of 0.776, 0.793, and 0.778 for fibrosis ≥F2, 0.788, 0.814, and 0.79 for fibrosis ≥F3, and 0.846, 0.859, and 0.856 for fibrosis ≥F4. The APRI consistently had the highest AUC values, except in individuals older than 64 years for fibrosis ≥F4. CONCLUSIONS The APRI was the most effective noninvasive fibrosis marker across NAFLD, MAFLD, and MASLD, particularly in age-stratified analyses. Further research is needed to establish standardized cut-off values and enhance the clinical utility of these markers in managing liver fibrosis.
Collapse
Affiliation(s)
- Se Young Jang
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Ki Tae Yoon
- Department of Internal Medicine, College of Medicine, Pusan National University, Busan, South Korea
- Liver Center, Pusan National University Yangsan Hospital, Yangsan-si, Gyeongsangnam-do, South Korea
| | - Young Youn Cho
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, South Korea
| | - Hoon Gil Jo
- Division of Gastroenterology, Department of Internal Medicine, Wonkwang University College of Medicine and Hospital, Iksan-si, Jeonlabuk-do, South Korea
| | - Yang Hyun Baek
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, South Korea
| | - Sang Yi Moon
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, South Korea
| | - Ae Jeong Jo
- Department of Information Statistics, Andong National University, Andong-si, Gyeongsangbuk-do, South Korea
| | - Young-Oh Kweon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Soo Young Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Yu Rim Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, South Korea
| | - Won Young Tak
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| |
Collapse
|
2
|
Rieder F, Nagy LE, Maher TM, Distler JHW, Kramann R, Hinz B, Prunotto M. Fibrosis: cross-organ biology and pathways to development of innovative drugs. Nat Rev Drug Discov 2025:10.1038/s41573-025-01158-9. [PMID: 40102636 DOI: 10.1038/s41573-025-01158-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/20/2025]
Abstract
Fibrosis is a pathophysiological mechanism involved in chronic and progressive diseases that results in excessive tissue scarring. Diseases associated with fibrosis include metabolic dysfunction-associated steatohepatitis (MASH), inflammatory bowel diseases (IBDs), chronic kidney disease (CKD), idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc), which are collectively responsible for substantial morbidity and mortality. Although a few drugs with direct antifibrotic activity are approved for pulmonary fibrosis and considerable progress has been made in the understanding of mechanisms of fibrosis, translation of this knowledge into effective therapies continues to be limited and challenging. With the aim of assisting developers of novel antifibrotic drugs, this Review integrates viewpoints of biologists and physician-scientists on core pathways involved in fibrosis across organs, as well as on specific characteristics and approaches to assess therapeutic interventions for fibrotic diseases of the lung, gut, kidney, skin and liver. This discussion is used as a basis to propose strategies to improve the translation of potential antifibrotic therapies.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA.
- Program for Global Translational Inflammatory Bowel Diseases (GRID), Chicago, IL, USA.
| | - Laura E Nagy
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Toby M Maher
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- National Heart and Lung Institute, Imperial College, London, UK
| | - Jörg H W Distler
- Department of Rheumatology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
- Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, RWTH Aachen; Medical Faculty, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
3
|
Nguyen MT, Lian A, Guilford FT, Venketaraman V. A Literature Review of Glutathione Therapy in Ameliorating Hepatic Dysfunction in Non-Alcoholic Fatty Liver Disease. Biomedicines 2025; 13:644. [PMID: 40149620 PMCID: PMC11940638 DOI: 10.3390/biomedicines13030644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global cause of liver dysfunction. This spectrum of hepatic disorders can progress to severe conditions, such as non-alcoholic steatohepatitis (NASH) and cirrhosis, due to oxidative stress and sustained cellular injury. With limited pharmacological options, glutathione (GSH), a key antioxidant, has shown promising potential in reducing oxidative stress, maintaining redox balance, and improving liver function. This literature review examines studies from 2014-2024 exploring GSH therapy in NAFLD patients. Eligible studies assessed GSH as the primary intervention for NAFLD in human subjects, reporting outcomes such as liver function or oxidative stress markers. Randomized clinical trials (RCTs) were eligible, while combination therapy studies were included if GSH's effect could be isolated. Exclusions applied to non-NAFLD studies, animal/in vitro models, and non-GSH antioxidant interventions. Analysis of three studies (totaling 109 participants) demonstrated consistent improvements in alanine transaminase (ALT) levels and reductions in oxidative stress markers like 8-hydroxy-2-deoxyguanosine (8-OHdG). However, small sample sizes and inconsistent protocols limit generalizability. Further large-scale RCTs are required to confirm GSH's efficacy, determine optimal dosing, and assess long-term effects. This literature review highlights GSH's potential as a novel NAFLD therapeutic strategy while emphasizing the need for further studies to refine its clinical application.
Collapse
Affiliation(s)
- Michelle Thuy Nguyen
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.T.N.); (A.L.)
| |
Collapse
|
4
|
Dell T, Mesropyan N, Layer Y, Tischler V, Weinhold L, Chang J, Jansen C, Schmidt B, Jürgens M, Isaak A, Kupczyk P, Pieper CC, Meyer C, Luetkens J, Kuetting D. Photon-counting CT-derived Quantification of Hepatic Fat Fraction: A Clinical Validation Study. Radiology 2025; 314:e241677. [PMID: 40100026 DOI: 10.1148/radiol.241677] [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: 03/20/2025]
Abstract
Background Steatosis is a critical health problem, creating a growing need for opportunistic screening. Early detection may allow for effective treatment and prevention of further liver complications. Purpose To evaluate photon-counting CT (PCCT) fat quantification on contrast-enhanced scans and validate the results against fat quantification via histopathologic assessment, controlled attenuation parameter (CAP) from transient elastography, and MRI proton density fat fraction (PDFF). Materials and Methods In this prospective, observational clinical study, PCCT-derived fat fraction quantification was assessed in participants with known or suspected liver disease. Participants underwent PCCT between February 2022 and January 2024. Participants also underwent biopsy, US with CAP measurement, or MRI with a PDFF sequence for hepatic fat fraction quantification. Liver fat fraction was measured on virtual noncontrast PCCT images using spectral processing software with a three-material decomposition algorithm for fat, liver tissue, and iodine. Steatosis was graded for each modality. Correlation between PCCT-based steatosis grades and biopsy- and CAP-based grades was assessed with the Spearman correlation coefficient. Agreement between PCCT and MRI PDFF measurements was assessed with the intraclass correlation coefficient. Receiver operating characteristic curve analysis was conducted to determine the optimal PCCT fat fraction threshold for distinguishing between participants with and those without steatosis. Results The study included 178 participants, of whom 27 (mean age, 60.7 years ± 15.2 [SD]; 18 male participants) underwent liver biopsy, 26 (mean age, 60.0 years ± 18.3; 15 male participants) underwent CAP measurement, and 125 (mean age, 61.2 years ± 13.1; 70 male participants) underwent MRI PDFF measurement. There was excellent agreement between PCCT and MRI PDFF assessment of liver fat fraction (intraclass correlation coefficient, 0.91 [95% CI: 0.87, 0.94]). In stratified analysis, the intraclass correlation coefficient was 0.84 (95% CI: 0.63, 0.93) in participants with known fibrosis and 0.92 (95% CI: 0.88, 0.94) in participants without fibrosis. There was moderate correlation of PCCT-based steatosis grade with histologic (ρ = 0.65) and CAP-based (ρ = 0.45) steatosis grade. Based on the Youden index, the PCCT fat fraction threshold that best discriminated between participants with and those without steatosis was 4.8%, with a maximum achievable sensitivity of 81% (38 of 47) and a specificity of 71% (55 of 78). Conclusion PCCT in a standard clinical setting allowed for accurate estimation of liver fat fraction compared with MRI PDFF-based reference standard measurements. © RSNA, 2025 See also the editorial by Kartalis and Grigoriadis in this issue.
Collapse
Affiliation(s)
- Tatjana Dell
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Narine Mesropyan
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Yannik Layer
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Verena Tischler
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Leonie Weinhold
- Institute for Medical Biometry, Informatics, and Epidemiology, Rhenish Friedrich Wilhelm University of Bonn, Bonn, Germany
| | - Johannes Chang
- Department of Internal Medicine I, Center for Cirrhosis and Portal Hypertension Bonn, University Hospital Bonn, Bonn, Germany
| | - Christian Jansen
- Department of Internal Medicine I, Center for Cirrhosis and Portal Hypertension Bonn, University Hospital Bonn, Bonn, Germany
| | - Bernhard Schmidt
- Department of Computed Tomography, Siemens Healthcare, Forchheim, Germany
| | - Markus Jürgens
- Department of Computed Tomography, Siemens Healthcare, Forchheim, Germany
| | - Alexander Isaak
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Patrick Kupczyk
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Claus Christian Pieper
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Carsten Meyer
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Julian Luetkens
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniel Kuetting
- Department of Diagnostic and Interventional Radiology and Quantitative Imaging Lab Bonn, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
5
|
Harvey BE. How improvements in US FDA regulatory process and procedures led to the drug approval for first ever treatment of a common liver disease. Acta Pharmacol Sin 2025; 46:515-524. [PMID: 39511464 PMCID: PMC11845757 DOI: 10.1038/s41401-024-01396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/13/2024] [Indexed: 11/15/2024]
Abstract
Metabolic-associated liver disease is a growing public health crisis, with phenotypes from fatty liver to steatohepatitis, previously known as NASH (Non-Alcoholic SteatoHepatitis) and currently rebranded as MASH (Metabolic dysfunction-Associated SteatoHepatitis). Dysfunction in liver metabolism can progress to liver fibrosis, end stage cirrhosis and death. MASH (NASH) is associated with an increased risk of cardiovascular disease, elevation in serum lipids and Type 2 Diabetes Mellitus. There is now a US-approved drug to treat patients with NASH (MASH) under the FDA Accelerated Approval Pathway, which requires follow-up outcome studies to confirm clinical benefit or risk drug withdrawal by the agency. Despite extra-hepatic factors that contribute to MASH and complicate clinical trial design, reorganization of the FDA drug premarket review divisions, improvements to agency policies and procedures, as well as updates to the US Food, Drug & Cosmetic Act (FD&C Act) upon which FDA regulation is based, have provided new agency tools that facilitated such a drug approval to address the profound unmet medical need for patients with this metabolic-based liver disease. There is reason for hope that continued evolution of the regulatory process will lead to additional drug approvals, as well as the ability for clinical trial endpoints studying MASH treatments to include both liver-based and traditional metabolic measures, independent of the specific FDA review division. This initial NASH/MASH FDA approval has also opened the door for initiation of Combination Clinical Trials, where the approved drug is paired with an experimental drug with a different mechanism of action, to increase overall efficacy and potentially minimize risks. It is envisioned that future treatment of NASH/MASH will mirror what is currently observed with Type 2 Diabetes Mellitus practice patterns, where multiple drugs with different mechanisms of actions are used to optimize treatment benefit/risk in the selected patient populations.
Collapse
|
6
|
Malandris K, Katsoula A, Liakos A, Karagiannis T, Sinakos E, Giouleme O, Klonizakis P, Theocharidou E, Gigi E, Bekiari E, Tsapas A. Diagnostic accuracy of Agile-4 score for liver cirrhosis in patients with metabolic dysfunction-associated steatotic liver disease. A systematic review and meta-analysis of diagnostic test accuracy studies. Diabetes Obes Metab 2025; 27:1406-1414. [PMID: 39703127 PMCID: PMC11802403 DOI: 10.1111/dom.16142] [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: 09/30/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
AIMS A novel noninvasive score, Agile-4 score, combining liver stiffness measurements, aspartate aminotransferase/alanine aminotransferase, platelet count, diabetes status and sex has been developed for the identification of cirrhosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). We assessed the performance of Agile-4 for ruling-in/out liver cirrhosis in MASLD patients. MATERIALS AND METHODS We searched Medline, Cochrane library, Web of science, Scopus and Echosens website up to May 2024. Eligible studies assessed the accuracy of Agile-4 for ruling-in (≥0.565) and ruling-out (<0.251) liver cirrhosis, using biopsy as the reference standard, at predefined thresholds. We calculated pooled sensitivity and specificity estimates for both Agile-4 thresholds alongside 95% confidence intervals following bivariate random-effect models. We assessed the risk of bias using Quality Assessment of Diagnostic Accuracy Studies-2 tool. RESULTS We included seven studies with 6037 participants. An Agile-4 score ≥0.565 yielded a pooled specificity of 0.93 (95% CI, 0.86-0.97). Similarly, an Agile-4 score <0.251 excluded cirrhosis with a summary sensitivity of 0.90 (0.80-0.95). Assuming a cirrhosis prevalence of 30%, the positive predictive value (PPV) for ruling-in cirrhosis was 80%, while the negative predictive value for ruling-out cirrhosis was 95%. Most studies were at high or unclear risk for bias due to concerns regarding patient selection and the blinding status of Agile-4 score interpretation in relation to biopsy results. CONCLUSIONS Agile-4 score performs well for ruling-in/out liver cirrhosis in MASLD patients. Owing to the relatively low PPV, sequential application of the Agile-4 after fibrosis-4 index (FIB-4) testing might further enhance its performance.
Collapse
Affiliation(s)
- Konstantinos Malandris
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Anastasia Katsoula
- Second Propaedeutic Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Aris Liakos
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Thomas Karagiannis
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Emmanouil Sinakos
- Fourth Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Olga Giouleme
- Second Propaedeutic Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Philippos Klonizakis
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Eleni Theocharidou
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Eleni Gigi
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Eleni Bekiari
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
| | - Apostolos Tsapas
- Clinical Research and Evidence‐Based Medicine Unit, Second Medical DepartmentAristotle University of ThessalonikiThessalonikiGreece
- Harris Manchester CollegeUniversity of OxfordOxfordUK
| |
Collapse
|
7
|
Kjær MB, Jørgensen AG, Fjelstrup S, Dupont DM, Bus C, Eriksen PL, Thomsen KL, Risikesan J, Nielsen S, Wernberg CW, Lauridsen MM, Bugianesi E, Rosso C, Grønbæk H, Kjems J. Diagnosis and Staging of Metabolic Dysfunction-Associated Steatotic Liver Disease Using Biomarker-Directed Aptamer Panels. Biomolecules 2025; 15:255. [PMID: 40001558 PMCID: PMC11852711 DOI: 10.3390/biom15020255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) affects one-third of adults globally. Despite efforts to develop non-invasive diagnostic tools, liver biopsy remains the gold standard for diagnosing metabolic dysfunction-associated steatohepatitis (MASH) and assessing fibrosis. This study investigated RNA aptamer panels, selected using APTASHAPE technology, for non-invasive MASLD diagnosis and fibrosis stratification. Aptamer panels were selected in a cohort of individuals with MASLD (development cohort, n = 77) and tested in separate cohorts: one with MASLD (test cohort, n = 57) and one assessed for bariatric surgery (bariatric cohort, n = 62). A panel distinguishing MASLD without steatohepatitis from MASH accurately stratified individuals in the developmentcohort (AUC = 0.83) but failed in the test and bariatric cohorts. It did, however, distinguish healthy controls from individuals with MASLD, achieving an AUC of 0.72 in the test cohort. A panel for fibrosis stratification differentiated F0 from F3-4 fibrosis in the development cohort (AUC = 0.68) but not in other cohorts. Mass spectrometry identified five plasma proteins as potential targets of the discriminative aptamers, with complement factor H suggested as a novel MASLD biomarker. In conclusion, APTASHAPE shows promise as a non-invasive tool for diagnosing and staging MASLD and identifying associated plasma biomarkers.
Collapse
Affiliation(s)
- Mikkel B. Kjær
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark; (M.B.K.); (P.L.E.); (K.L.T.)
- Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark; (J.R.); (S.N.)
| | - Asger G. Jørgensen
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (A.G.J.); (S.F.); (D.M.D.); (C.B.)
| | - Søren Fjelstrup
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (A.G.J.); (S.F.); (D.M.D.); (C.B.)
| | - Daniel M. Dupont
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (A.G.J.); (S.F.); (D.M.D.); (C.B.)
| | - Claus Bus
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (A.G.J.); (S.F.); (D.M.D.); (C.B.)
| | - Peter L. Eriksen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark; (M.B.K.); (P.L.E.); (K.L.T.)
| | - Karen L. Thomsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark; (M.B.K.); (P.L.E.); (K.L.T.)
| | - Jeyanthini Risikesan
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark; (J.R.); (S.N.)
| | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark; (J.R.); (S.N.)
| | - Charlotte W. Wernberg
- Department of Gastroenterology and Hepatology, University Hospital of Southern Denmark, 6700 Esbjerg, Denmark; (C.W.W.); (M.M.L.)
- ATLAS Centre for Functional Genomics, University of Southern Denmark, 5230 Odense, Denmark
| | - Mette M. Lauridsen
- Department of Gastroenterology and Hepatology, University Hospital of Southern Denmark, 6700 Esbjerg, Denmark; (C.W.W.); (M.M.L.)
- ATLAS Centre for Functional Genomics, University of Southern Denmark, 5230 Odense, Denmark
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, Via Verdi 8, 10124 Torino, Italy; (E.B.); (C.R.)
| | - Chiara Rosso
- Department of Medical Sciences, University of Turin, Via Verdi 8, 10124 Torino, Italy; (E.B.); (C.R.)
| | - Henning Grønbæk
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark; (M.B.K.); (P.L.E.); (K.L.T.)
- Department of Clinical Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (A.G.J.); (S.F.); (D.M.D.); (C.B.)
| |
Collapse
|
8
|
Karin M, Kim JY. MASH as an emerging cause of hepatocellular carcinoma: current knowledge and future perspectives. Mol Oncol 2025; 19:275-294. [PMID: 38874196 PMCID: PMC11793012 DOI: 10.1002/1878-0261.13685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
Hepatocellular carcinoma is one of the deadliest and fastest-growing cancers. Among HCC etiologies, metabolic dysfunction-associated fatty liver disease (MAFLD) has served as a major HCC driver due to its great potential for increasing cirrhosis. The obesogenic environment fosters a positive energy balance and results in a continuous rise of obesity and metabolic syndrome. However, it is difficult to understand how metabolic complications lead to the poor prognosis of liver diseases and which molecular mechanisms are underpinning MAFLD-driven HCC development. Thus, suitable preclinical models that recapitulate human etiologies are essentially required. Numerous preclinical models have been created but not many mimicked anthropometric measures and the course of disease progression shown in the patients. Here we review the literature on adipose tissues, liver-related HCC etiologies and recently discovered genetic mutation signatures found in MAFLD-driven HCC patients. We also critically review current rodent models suggested for MAFLD-driven HCC study.
Collapse
Affiliation(s)
- Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Ju Youn Kim
- Department of Molecular and Life ScienceHanyang University ERICAAnsanKorea
| |
Collapse
|
9
|
Serdjebi C, Foucher J, Besson A, Gay J, Delamarre A, Cohen-Bacrie C. Hepatoscope 2DTE's image-based quality index enhances applicability and repeatability of liver stiffness measurement. Clin Res Hepatol Gastroenterol 2025; 49:102532. [PMID: 39832729 DOI: 10.1016/j.clinre.2025.102532] [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: 11/14/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE Hepatoscope® is an ultraportable ultrasound system with 50 Hz two-dimensional transient elastography (2DTE) for liver stiffness measurement (LSM). It provides a quality index (QI) for individual stiffness values that is based on imaging features. This study evaluated the 2DTE intra- and inter-user repeatability in patients with chronic liver diseases (CLD) for novice and expert operators across various QI conditions. We compared the performances with other imaging and non-imaging elastography techniques. METHODS This investigation was a prospective cross-sectional single-center study. One hundred CLD patients underwent LSMs with vibration-controlled transient elastography (VCTE™), two-dimensional shear wave elastography (2DSWE) and 2DTE. Expert and novice operators each performed two consecutive exams with 2DTE, blinded to any output. Intra-class correlation coefficient for intra-, and inter-user repeatability were calculated as well as applicability for various QI conditions, and Spearman's correlations against VCTE and 2DSWE were assessed. RESULTS Computing LSM as the median of 10 stiffness values with a QI>85 % yielded the best compromise between reliability and applicability. In this setting, expert and inter-operator repeatability showed significant improvement over the condition with no QI threshold (ICC = 0.81 and 0.79, respectively) and 95 % applicability. Novice repeatability was excellent with a more restrictive QI (ICC = 0.80). LSM with 2DTE demonstrated strong and moderate correlation with VCTE and 2DSWE (r = 0.63 and 0.55, respectively). CONCLUSION Hepatoscope 2DTE provides a promising and reliable alternative for non-invasive liver stiffness assessment in patients with CLD. The application of the image-based QI enhances LSM reliability and consistency across novice and expert operators.
Collapse
Affiliation(s)
| | | | | | - Joel Gay
- E-Scopics, Aix-en-provence, France
| | - Adèle Delamarre
- Bordeaux Institute of Oncology, INSERM U1312, University of Bordeaux, France
| | | |
Collapse
|
10
|
Stefan N, Yki-Järvinen H, Neuschwander-Tetri BA. Metabolic dysfunction-associated steatotic liver disease: heterogeneous pathomechanisms and effectiveness of metabolism-based treatment. Lancet Diabetes Endocrinol 2025; 13:134-148. [PMID: 39681121 DOI: 10.1016/s2213-8587(24)00318-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 12/18/2024]
Abstract
The global epidemic of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing worldwide. People with MASLD can progress to cirrhosis and hepatocellular carcinoma and are at increased risk of developing type 2 diabetes, cardiovascular disease, chronic kidney disease, and extrahepatic cancers. Most people with MASLD die from cardiac-related causes. This outcome is attributed to the shared pathogenesis of MASLD and cardiometabolic diseases, involving unhealthy dietary habits, dysfunctional adipose tissue, insulin resistance, and subclinical inflammation. In addition, the steatotic and inflamed liver affects the vasculature and heart via increased glucose production and release of procoagulant factors, dyslipidaemia, and dysregulated release of hepatokines and microRNAs. However, there is substantial heterogeneity in the contributors to the pathophysiology of MASLD, which might influence its rate of progression, its relationship with cardiometabolic diseases, and the response to therapy. The most effective non-pharmacological treatment approaches for people with MASLD include weight loss. Paradoxically, some effective pharmacological approaches to improve liver health in people with MASLD are associated with no change in bodyweight or even with weight gain, and similar response heterogeneity has been observed for changes in cardiometabolic risk factors. In this Review, we address the heterogeneity of MASLD with respect to its pathogenesis, outcomes, and metabolism-based treatment responses. Although there is currently insufficient evidence for the implementation of precision medicine for risk prediction, prevention, and treatment of MASLD, we discuss whether knowledge about this heterogeneity might help achieving this goal in the future.
Collapse
Affiliation(s)
- Norbert Stefan
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich, Tübingen, Germany; German Center for Diabetes Research, Neuherberg, Germany.
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | |
Collapse
|
11
|
Yang W, Yan X, Chen R, Xin X, Ge S, Zhao Y, Yan X, Zhang J. Smad4 deficiency in hepatocytes attenuates NAFLD progression via inhibition of lipogenesis and macrophage polarization. Cell Death Dis 2025; 16:58. [PMID: 39890803 PMCID: PMC11785999 DOI: 10.1038/s41419-025-07376-8] [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: 05/31/2024] [Revised: 12/22/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a major cause of chronic liver disorders, has become a serious public health issue. Although the Smad4 signaling pathway has been implicated in the progression of NAFLD, the specific role of Smad4 in hepatocytes in NAFLD pathogenesis remains unclear. Hepatocyte-specific knockout Smad4 mice (AlbSmad4-/-) were first constructed using the Cre-Loxp recombinant system to establish a high-fat diet induced NAFLD model. The role of Smad4 in the occurrence and development of NAFLD was determined by monitoring the body weight of mice, detecting triglycerides and free fatty acids in serum and liver tissue homogenates, staining the tissue sections to observe the accumulation of liver fat, and RT-qPCR detecting the expression of genes related to lipogenesis, fatty acid intake, and fatty acid β oxidation. The molecular mechanism of Smad4 in hepatocytes affecting NAFLD was therefore investigated through combining in vitro and in vivo experiments. Smad4 deficiency in hepatocytes mitigated NAFLD progression and decreased inflammatory cell infiltration. Moreover, Smad4 deficiency inhibited CXCL1 secretion by suppressing the activation of the ASK1/P38/JNK signaling pathway. Furthermore, targeting CXCL1 using CXCR2 inhibitors diminished hepatocyte lipogenesis and inhibited the polarization of M1-type macrophages. Collectively, these results suggested that Smad4 plays a vital role in exacerbating NAFLD and may be a promising candidate for anti-NAFLD therapy.
Collapse
Affiliation(s)
- Wei Yang
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targetubg Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Xuanxuan Yan
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Rui Chen
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Xin Xin
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Shuang Ge
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targetubg Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Yongxiang Zhao
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targetubg Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, China.
| | - Jinhua Zhang
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China.
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targetubg Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning, China.
| |
Collapse
|
12
|
García S, Monserrat-Mesquida M, Ugarriza L, Casares M, Gómez C, Mateos D, Angullo-Martínez E, Tur JA, Bouzas C. Ultra-Processed Food Consumption and Metabolic-Dysfunction-Associated Steatotic Liver Disease (MASLD): A Longitudinal and Sustainable Analysis. Nutrients 2025; 17:472. [PMID: 39940330 PMCID: PMC11820933 DOI: 10.3390/nu17030472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND The rising prevalence of metabolic-dysfunction-associated steatotic liver disease (MASLD) is a significant health challenge, and the consumption of ultra-processed foods (UPFs) could play a key role. AIM The aim is assess the impact of UPF consumption changes on the development and progression of MASLD in adults. DESIGN This is a longitudinal study to assess how changes in UPF consumption affect liver fat and MASLD parameters over 6 months in 70 participants. METHODS Dietary intake was assessed using a validated food frequency questionnaire, and foods were classified according to the NOVA system. Participants were divided into three groups based on UPF consumption changes: maximum (T1), medium (T2), and minimum reduction (T3). Fatty liver parameters were assessed with magnetic resonance imaging and ultrasonography. Mediterranean diet (Med-diet) adherence and sociodemographic parameters were also recorded. The General Linear Model was used to determine relationships between UPF consumption, fatty liver disease parameters, and diet. RESULTS Participants in T1 experienced a 7.7% reduction in intrahepatic fat content (IFC) compared to 2.6% in T3. T1 showed increased Med-diet adherence and decreased meat and sweets consumption. The energy intake decreased by 605.3 kcal/day in T1, while T3 showed an increase of 209.5 kcal/day. CONCLUSIONS Reducing UPF consumption leads to a decrease in IFC, associated with high Med-diet adherence and low calorie intake. Adopting these dietary patterns aligns with global sustainability goals and could further benefit MASLD patients by addressing environmental challenges alongside improving liver health.
Collapse
Affiliation(s)
- Silvia García
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Margalida Monserrat-Mesquida
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Lucía Ugarriza
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
- Primary Health Care Center Camp Redó, IBSalut, 07010 Palma de Mallorca, Spain
| | - Miguel Casares
- Radiodiagnosis Service, Red Asistencial Juaneda, 07011 Palma de Mallorca, Spain
| | - Cristina Gómez
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- Clinical Analysis Service, University Hospital Son Espases, 07120 Palma de Mallorca, Spain
| | - David Mateos
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
- Hospital of Manacor, 07500 Manacor, Spain
| | - Escarlata Angullo-Martínez
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
- Primary Health Care Center Escola Graduada, IBSalut, 07001 Palma de Mallorca, Spain
| | - Josep A. Tur
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Cristina Bouzas
- Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma de Mallorca, Spain; (S.G.); (M.M.-M.); (C.G.); (C.B.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
13
|
Lu C, Han M, Ma Q, Ying L, Zhang Y. Identification of biomarkers associated with coronary artery disease and non-alcoholic fatty liver disease by bioinformatics analysis and machine learning. Sci Rep 2025; 15:3557. [PMID: 39875572 PMCID: PMC11775188 DOI: 10.1038/s41598-025-87923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
The constantly emerging evidence indicates a close association between coronary artery disease (CAD) and non-alcoholic fatty liver disease (NAFLD). However, the exact mechanisms underlying their mutual relationship remain undefined. This study aims to explore the common signature genes, potential mechanisms, diagnostic markers, and therapeutic targets for CAD and NAFLD. We downloaded CAD and NAFLD datasets from the Gene Expression Omnibus (GEO) database and analyzed the differentially expressed genes (DEGs) by limma. Protein-protein interaction (PPI) network was constructed with common DEGs (co-DEGs), and hub genes were screened by Maximal Clique Centrality (MCC) algorithm. Candidate biomarkers were selected from intersection of three machine learning algorithms. Expression levels, nomogram, the areas under the receiver operating characteristic curve (AUC) of candidate biomarkers were performed. CIBERSORT algorithm was used to assess the immune cell infiltration, and Spearman's correlations tests were used for calculating the correlation of biomarker genes. A total of 554 overlapping DEGs associated with CAD and NAFLD were obtained by analysis of GSE113079 and GSE89632 datasets. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that the co-DEGs were significantly enriched in immune effector process, inflammation response and lipid metabolism. The PPI network generated a 1245-edge network, and top 50 genes were selected using the MCC algorithm. The candidate biomarkers were screened from intersection of machine learning in GSE89632, including CEBPA, CXCL2, JUN and FOXO1. The ROC results showed that these four biomarker genes have good diagnostic value for patients with both CAD and NAFLD. Then we explored the immune landscape, immune infiltration and the correlation between biomarker gene expression in CAD and NAFLD samples. In this study, we predict that CEBPA, CXCL2, JUN and FOXO1 can be used to diagnose CAD and NAFLD. Our study provided new insights for potential biomarkers, molecular mechanism and therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Chuan Lu
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, 116021, China
| | - Mei Han
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China
| | - Qiqi Ma
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China
| | - Li Ying
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China.
| | - Yue Zhang
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China.
| |
Collapse
|
14
|
Harrison SA, Browne SK, Suschak JJ, Tomah S, Gutierrez JA, Yang J, Roberts MS, Harris MS. Effect of pemvidutide, a GLP-1/glucagon dual receptor agonist, on MASLD: A randomized, double-blind, placebo-controlled study. J Hepatol 2025; 82:7-17. [PMID: 39002641 DOI: 10.1016/j.jhep.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND & AIMS This was a randomized, double-blind, placebo-controlled study to assess the effects of pemvidutide, a glucagon-like peptide-1 (GLP-1)/glucagon dual receptor agonist, on liver fat content (LFC) in individuals with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Patients with a BMI ≥28.0 kg/m2 and LFC ≥10% by magnetic resonance imaging-proton density fat fraction were randomized 1:1:1:1 to pemvidutide at 1.2 mg, 1.8 mg, or 2.4 mg, or placebo administered subcutaneously once weekly for 12 weeks. Participants were stratified according to a diagnosis of type 2 diabetes mellitus. The primary efficacy endpoint was relative reduction (%) from baseline in LFC after 12 weeks of treatment. RESULTS Ninety-four patients were randomized and dosed. Median baseline BMI and LFC across the study population were 36.2 kg/m2 and 20.6%; 29% of patients had type 2 diabetes mellitus. At week 12, relative reductions in LFC from baseline were 46.6% (95% CI -63.7 to -29.6), 68.5% (95% CI -84.4 to -52.5), and 57.1% (95% CI -76.1 to -38.1) for the pemvidutide 1.2 mg, 1.8 mg, and 2.4 mg groups, respectively, vs. 4.4% (95% CI -20.2 to 11.3) for the placebo group (p <0.001 vs. placebo, all treatment groups), with 94.4% and 72.2% of patients achieving 30% and 50% reductions in LFC and 55.6% achieving normalization (≤5% LFC) at the 1.8 mg dose. Maximal responses for weight loss (-4.3%; p <0.001), alanine aminotransferase (-13.8 IU/L; p = 0.029), and corrected cT1 (-75.9 ms; p = 0.002) were all observed at the 1.8 mg dose. Pemvidutide was well-tolerated at all doses with no severe or serious adverse events. CONCLUSIONS In patients with MASLD, weekly pemvidutide treatment yielded significant reductions in LFC, markers of hepatic inflammation, and body weight compared to placebo. IMPACT AND IMPLICATIONS Metabolic dysfunction-associated steatotic liver disease, and its progressive form steatohepatitis, are strongly associated with overweight/obesity and it is believed that the excess liver fat associated with obesity is an important driver of these diseases. Glucagon-like peptide-1 receptor (GLP-1R) agonists elicit weight loss through centrally and peripherally mediated effects on appetite. Unlike GLP-1R agonists, glucagon receptor agonists act directly on the liver to stimulate fatty acid oxidation and inhibit lipogenesis, potentially providing a more potent mechanism for liver fat content reduction than weight loss alone. This study demonstrated the ability of once-weekly treatment with pemvidutide, a dual GLP-1R/glucagon receptor agonist, to significantly reduce liver fat content, hepatic inflammatory activity, and body weight, suggesting that pemvidutide may be an effective treatment for both metabolic dysfunction-associated steatohepatitis and obesity. CLINICAL TRIAL NUMBER NCT05006885.
Collapse
Affiliation(s)
- Stephen A Harrison
- Department of Hepatology, University of Oxford, Oxford, UK; Pinnacle Clinical Research, San Antonio, TX, USA
| | | | | | | | - Julio A Gutierrez
- Altimmune, Inc, Gaithersburg, MD, USA; Center for Organ Transplant, Scripps, La Jolla, Ca, USA
| | - Jay Yang
- Altimmune, Inc, Gaithersburg, MD, USA
| | | | | |
Collapse
|
15
|
Lassailly G, Caiazzo R, Goemans A, Chetboun M, Gnemmi V, Labreuche J, Baud G, Verkindt H, Marciniak C, Oukhouya-Daoud N, Ntandja-Wandji LC, Ningarhari M, Leteurtre E, Raverdy V, Dharancy S, Louvet A, Pattou F, Mathurin P. Resolution of Metabolic Dysfunction-associated Steatohepatitis With No Worsening of Fibrosis After Bariatric Surgery Improves 15-year Survival: A Prospective Cohort Study. Clin Gastroenterol Hepatol 2024:S1542-3565(24)01078-4. [PMID: 39709138 DOI: 10.1016/j.cgh.2024.10.025] [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: 08/15/2024] [Revised: 10/01/2024] [Accepted: 10/20/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND & AIMS The aim of this study was to investigate the consequences of the histological progression of metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis on long-term survival after bariatric surgery. METHODS From 1994 to 2021, 3028 patients at the University Hospital of Lille were prospectively included. Baseline liver biopsies were systematically performed with proposed follow-up biopsies 1 year after surgery, mainly in patients with MASH. We evaluated the association of the baseline and 1-year histologic progression of MASH and fibrosis status and long-term survival using Cox regression models. RESULTS At baseline, 2641 patients (89%) had a biopsy, including 232 with MASH (8.7%) and 266 (10.8%) with significant fibrosis (grade F2-F4). The median follow-up was 10.1 years. At 1 year, 594 patients had qualitative paired biopsies. Survival was shorter at the 15-year follow-up in patients with baseline MASH, than in those without (hazard ratio [HR], 2.21; 95% confidence interval [CI], 1.38-3.53) and in F2 to F4 than in F0 to F1 (HR, 3.38; 95% CI, 2.24-5.10). At the 1-year landmark analysis, compared with patients without baseline MASH, mortality increased in those with persistent MASH and/or if fibrosis worsened (adjusted HR, 2.54; 95% CI, 1.06-6.10), but not if MASH resolved without the worsening of fibrosis (adjusted HR, 0.73; 95% CI, 0.28-1.87). Similarly, compared with patients without significant fibrosis at baseline, patients with persistent significant fibrosis had increased mortality (adjusted HR, 4.03; 95% CI, 1.86-8.72) but not if fibrosis improved from F2 to F4 to F0 to F1 (adjusted HR; 1.49; 95% CI, 0.52-4.24). CONCLUSION Histologic remission of MASH or significant fibrosis improves survival after bariatric surgery.
Collapse
Affiliation(s)
- Guillaume Lassailly
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Robert Caiazzo
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Armelle Goemans
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Mikael Chetboun
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Viviane Gnemmi
- Service d'Anatomopathologie, Centre Hospitalier Universitaire de Lille, Université de Lille, INSERM UMR-S 1172, Lille, France
| | | | - Gregory Baud
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Helene Verkindt
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Camille Marciniak
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Naima Oukhouya-Daoud
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Line-Carolle Ntandja-Wandji
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Massih Ningarhari
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Emmanuelle Leteurtre
- Service d'Anatomopathologie, Centre Hospitalier Universitaire de Lille, Université de Lille, INSERM UMR-S 1172, Lille, France
| | - Violeta Raverdy
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Sébastien Dharancy
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Alexandre Louvet
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - François Pattou
- Service de Chirurgie Générale et Endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France.
| | - Philippe Mathurin
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France.
| |
Collapse
|
16
|
Lahooti A, Westerveld D, Johnson K, Aneke-Nash C, Baig MU, Akagbosu C, Hanscom M, Buckholz A, Newberry C, Herr A, Schwartz R, Yeung M, Sampath K, Mahadev S, Kumar S, Carr-Locke D, Aronne L, Shukla A, Sharaiha RZ. Improvement in obesity-related comorbidities 5 years after endoscopic sleeve gastroplasty: a prospective cohort study. Gastrointest Endosc 2024:S0016-5107(24)03792-1. [PMID: 39694295 DOI: 10.1016/j.gie.2024.12.017] [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: 08/30/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND AND AIMS Endoscopic sleeve gastroplasty (ESG) is a minimally invasive bariatric procedure of the gastric cavity to facilitate weight loss. We aimed to evaluate the long-term effects of ESG as a monotherapy on obesity-related comorbidities over 5 years. METHODS This prospective study analyzed data from 404 consecutive patients (aged 45 ± 11.9 years, 76% women) who underwent ESG from August 2013 through June 2024. All patients had body mass indices ≥30 or ≥27 kg/m2 with comorbidities. Patients receiving adjuvant therapy were excluded to assess ESG as a monotherapy. All procedures were performed with a flexible endoscopic suturing system to facilitate restriction of the stomach. Patients were evaluated after 12 months (n = 336), 36 months (n = 210), and 60 months (n = 196) for anthropometric features, clinical parameters, metabolic panels, and liver function tests. Primary outcomes were the effects of ESG on obesity-related comorbidities and metabolic biomarkers. Secondary outcomes were total body weight loss (TBWL) and safety. RESULTS At 5 years after ESG, patients had significant reductions in levels of hemoglobin A1c (Δ .42%, P = .0007), systolic blood pressure (Δ 4.1 mm Hg, P = .0071), and alanine aminotransferase (Δ 7.4 U/L, P = .002). Body mass indices before the procedure were 37.5 ± 5.8 and 33.8 ± 6.7 kg/m2 at 60 months (P < .001). TBWL was 13.4% at 12 months (84.5% follow-up), 9.9% at 36 months (63.3% follow-up), and 11.8% at 60 months (74.8% follow-up), with 3 moderate adverse events (.7%). CONCLUSIONS ESG is an effective, noninvasive monotherapy that improved markers of hypertension, diabetes, and metabolic dysfunction-associated steatotic liver disease and led to a TBWL of 11.8% at 5 years after the procedure. (Clinical trial registration number: NCT04494048.).
Collapse
Affiliation(s)
- Ali Lahooti
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Donevan Westerveld
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kate Johnson
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Chino Aneke-Nash
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Muhammad Usman Baig
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Cynthia Akagbosu
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Mark Hanscom
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Adam Buckholz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Carolyn Newberry
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Andrea Herr
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Robert Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Michele Yeung
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Kartik Sampath
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Srihari Mahadev
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Sonal Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - David Carr-Locke
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Louis Aronne
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Alpana Shukla
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Reem Z Sharaiha
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
17
|
Iwaki M, Kobayashi T, Nogami A, Ogawa Y, Imajo K, Sakai E, Nakada Y, Koyama S, Kurihashi T, Oza N, Kohira T, Okada M, Yamaguchi Y, Iwane S, Kageyama F, Sasada Y, Matsushita M, Tadauchi A, Murohisa G, Nagasawa M, Sato S, Maeda K, Furuta K, Shigefuku R, Seko Y, Tobita H, Kawata K, Kawanaka M, Sugihara T, Tamaki N, Iwasa M, Kawaguchi T, Itoh Y, Kawaguchi A, Takahashi H, Nakajima A, Yoneda M. Pemafibrate for treating MASLD complicated by hypertriglyceridaemia: a multicentre, open-label, randomised controlled trial study protocol. BMJ Open 2024; 14:e088862. [PMID: 39581726 PMCID: PMC11590823 DOI: 10.1136/bmjopen-2024-088862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease, now known as metabolic dysfunction-associated steatotic liver disease (MASLD), is a phenotype of the metabolic syndrome in the liver and is clearly associated with metabolic abnormalities such as hyperglycaemia and dyslipidaemia. Although the prevalence of MASLD is increasing worldwide, there is currently no consensus on the efficacy and safety of the drugs used to treat MASLD/metabolic dysfunction-associated steatohepatitis (MASH). Pemafibrate, a selective peroxisome proliferator-activated receptor alpha modulator, was designed to have higher peroxisome proliferator-activated receptor alfa (PPARα) agonist activity and selectivity than existing PPARα agonists, and in development trials, without increasing creatinine levels, lipid parameters and alanine aminotransferase (ALT) were significantly improved. Thus, pemafibrate may effectively ameliorate the pathogenesis and metabolic abnormalities in MASLD/MASH. In this trial, we evaluated the efficacy and safety of pemafibrate in patients with MASLD/MASH. METHODS AND ANALYSIS This trial was designed as an open-label, three-arm, randomised controlled study. After obtaining informed consent, patients aged 20-80 years who met the selection criteria were enrolled. Patients were randomised to receive pemafibrate 0.4 mg/day, 0.2 mg/day or fenofibrate (n=120 per group). The duration of treatment was 48 weeks. The primary endpoint was a change in ALT levels after 24 weeks of administration. Secondary endpoints included changes from baseline in liver fibrosis markers (fibrosis-4 index, type IV collagen 7s, enhanced liver fibrosis and Mac-2 binding protein glycosylation isomer) at 48 weeks as well as changes in liver fat mass and liver stiffness measured by MRI and ultrasound (US) at centres equipped with MRI and US capabilities. ETHICS AND DISSEMINATION Ethical approval was obtained from the Yokohama City University Certified Institutional Review Board before participant enrolment (CRB20-014). The results of this study will be submitted for publication in international peer-reviewed journals and the key findings will be presented at international scientific conferences. Participants wishing to understand the results of this study will be contacted directly on data publication. TRIAL REGISTRATION NUMBER This trial was registered in the Japan Registry of Clinical Trials (number: jRCTs031200280). PROTOCOL VERSION V.1.9, 23 November 2023.
Collapse
Affiliation(s)
- Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yuji Ogawa
- Department of Gastroenterology, National Hospital Organisation Yokohama Medical Center, Yokohama, Japan
| | - Kento Imajo
- Department of Gastroenterology, Shin Yurigaoka General Hospital, Kawasaki, Kanagawa, Japan
| | - Eiji Sakai
- Department of Gastroenterology, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | - Yoshinobu Nakada
- Department of Internal Medicine, Shonan Hospital, Yokosuka, Kanagawa, Japan
| | - Satoshi Koyama
- Department of Internal Medicine, NamikiKoiso-Medical Clinic, Yokohama, Japan
| | - Takeo Kurihashi
- Department of Internal Medicine, Kanagawa Dental University Yokohama Clinic, Yokohama, Japan
| | - Noriko Oza
- Department of Hepato-Biliary-Pancreatology, Saga Prefecture Medical Center Koseikan, Saga, Saga, Japan
| | | | - Michiaki Okada
- Department of Internal Medicine, Karatsu Red Cross Hospital, Karatsu, Japan
| | - Yuki Yamaguchi
- Department of Internal Medicine, Masuda Red Cross Hospital, Masuda, Japan
| | - Shinji Iwane
- Department of Internal Medicine, Fujioka Hospital, Saga, Japan
| | - Fujito Kageyama
- Department of Gastroenterology and Hepatology, Hamamatsu Medical Center, Hamamatsu, Shizuoka, Japan
| | - Yuzo Sasada
- Division of Hepatology, Iwata City Hospital, Iwata, Japan
| | | | - Akimitsu Tadauchi
- Department of Gastroenterology, Seirei Mikatahara Byoin, Hamamatsu, Shizuoka, Japan
| | - Gou Murohisa
- Department of Gastroenterology, Seirei Hamamatsu Byoin, Hamamatsu, Shizuoka, Japan
| | - Masamichi Nagasawa
- Department of Gastroenterology, Seirei Hamamatsu Byoin, Hamamatsu, Shizuoka, Japan
| | - Shuichi Sato
- Department of Internal Medicine, Izumo City General Medical Center, Izumo, Japan
| | - Kazuhisa Maeda
- Department of Internal Medicine, Kitasenri Maeda Clinic, Suita, Japan
| | - Koichiro Furuta
- Department of Gastroenterology, National Hospital Organization Hamada Medical Center, Hamada, Japan
| | - Ryuta Shigefuku
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine Faculty of Medicine, Tsu, Mie, Japan
| | - Yuya Seko
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Tobita
- Division of Hepatology, Shimane University Hospital, Shimane, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, Kawasaki Medical Center, Kawasaki Medical School, Kurashiki, Japan
| | - Takaaki Sugihara
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Tottori University Faculty of Medicine Graduate School of Medicine, Yonago, Tottori, Japan
| | - Nobuharu Tamaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Musashino, Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine Faculty of Medicine, Tsu, Mie, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Kawaguchi
- Education and Research Center for Community Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirokazu Takahashi
- Department of Metabolism and Endocrinology, Liver Center, Saga University Hospital, Saga, Saga, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
18
|
Yan Y, Gan D, Zhang P, Zou H, Li M. A machine learning-based predictive model discriminates nonalcoholic steatohepatitis from nonalcoholic fatty liver disease. Heliyon 2024; 10:e38848. [PMID: 39512464 PMCID: PMC11539579 DOI: 10.1016/j.heliyon.2024.e38848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/31/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a leading cause of liver-related morbidity and mortality. The diagnosis of non-alcoholic steatohepatitis (NASH) plays a crucial role in the management of NAFLD patients. Objective The aim of our observational study was to build a machine learning model to identify NASH in NAFLD patients. Methods The clinical characteristics of 259 NAFLD patients and their initial laboratory data (Cohort 1) were collected to train the model and carry out internal validation. We compared the models built by five machine learning algorithms and screened out the best models. Receiver operating characteristic (ROC) curves, sensitivity, specificity, and accuracy were used to evaluate the performance of the model. In addition, the NAFLD patients in Cohort 2 (n = 181) were externally verified. Results We finally identified six independent risk factors for predicting NASH, including neutrophil percentage (NEU%), aspartate aminotransferase/alanine aminotransferase (AST/ALT), hematocrit (HCT), creatinine (CREA), uric acid (UA), and prealbumin (PA). The NASH-XGB6 model built using the XGBoost algorithm showed sufficient prediction accuracy, with ROC values of 0.95 (95 % CI, 0.91-0.98) and 0.90 (95 % CI, 0.88-0.93) in Cohort 1 and Cohort 2, respectively. Conclusions NASH-XGB6 can serve as an effective tool for distinguishing NASH patients from NAFLD patients.
Collapse
Affiliation(s)
- Yuqi Yan
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Danhui Gan
- Department of Clinical Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Ping Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Haizhu Zou
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - MinMin Li
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| |
Collapse
|
19
|
Harrison SA, Dubourg J. Liver biopsy evaluation in MASH drug development: Think thrice, act wise. J Hepatol 2024; 81:886-894. [PMID: 38879176 DOI: 10.1016/j.jhep.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/27/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024]
Abstract
During recent decades, the metabolic dysfunction-associated steatohepatitis (MASH) field has witnessed several paradigm shifts, including the recognition of liver fibrosis as the main predictor of major adverse liver outcomes. Throughout this evolution, liver histology has been recognised as one of the main hurdles in MASH drug development due to its invasive nature, associated cost, and high inter- and intra-reader variability. Collective experience demonstrates the importance of consistency in the central reading process, where consensus methods have emerged as appropriate ways to mitigate against well-known challenges. Using crystalized knowledge in the field, stakeholders should collectively work towards the next paradigm shift, where non-invasive biomarkers will be considered surrogate endpoints for accelerated approval. In this review, we provide an overview of the evolution of the regulatory histology endpoints and the liver biopsy reading process, within the MASH trial landscape, over recent decades; we then review the biggest challenges associated with liver biopsy endpoints. Finally, we discuss and provide recommendations on the best practices for liver biopsy evaluation in MASH drug development.
Collapse
Affiliation(s)
- Stephen A Harrison
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | | |
Collapse
|
20
|
Mallet M, Silaghi CA, Sultanik P, Conti F, Rudler M, Ratziu V, Thabut D, Pais R. Current challenges and future perspectives in treating patients with NAFLD-related cirrhosis. Hepatology 2024; 80:1270-1290. [PMID: 37183906 DOI: 10.1097/hep.0000000000000456] [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: 01/23/2023] [Accepted: 04/20/2023] [Indexed: 05/16/2023]
Abstract
Despite the slow, progressive nature of NAFLD, the number of patients with NAFLD-related cirrhosis has significantly increased. Although the management of patients with cirrhosis is constantly evolving, improving the prognosis of patients with NAFLD-related cirrhosis is a challenge because it is situated at the crossroads between the liver, the metabolic, and the cardiovascular diseases. Therefore, the therapeutic interventions should not only target the liver but also the associated cardiometabolic conditions and should be adapted accordingly. The objective of the current review is to critically discuss the particularities in the management of patients with NAFLD-related cirrhosis. We relied on the recommendations of scientific societies and discussed them in the specific context of NAFLD cirrhosis and the surrounding cardiometabolic milieu. Herein, we covered the following aspects: (1) the weight loss strategies through lifestyle interventions to avoid sarcopenia and improve portal hypertension; (2) the optimal control of metabolic comorbidities in particular type 2 diabetes aimed not only to improve cardiovascular morbidity/mortality but also to lower the incidence of cirrhosis-related complications (we discussed various aspects related to the safety of oral antidiabetic drugs in cirrhosis); (3) the challenges in performing bariatric surgery in patients with cirrhosis related to the portal hypertension and the risk of cirrhosis decompensation; (4) the particularities in the diagnosis and management of the portal hypertension and the difficulties in managing patients awaiting for liver transplantation; and (5) the difficulties in developing drugs and conducting clinical trials in patients with NAFLD-related cirrhosis. Moreover, we discussed the emerging options to overcome these obstacles.
Collapse
Affiliation(s)
- Maxime Mallet
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Cristina Alina Silaghi
- Department of Endocrinology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Roumanie
| | - Philippe Sultanik
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS), Paris, France
| | - Filomena Conti
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
| | - Marika Rudler
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS), Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Vlad Ratziu
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
- INSERM UMRS 1138 CRC, Paris, France
| | - Dominique Thabut
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Brain Liver Pitié-Salpêtrière Study Group (BLIPS), Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
| | - Raluca Pais
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Service d'hepato-gastroentérologie, Hôpital Pitié-Salpêtrière, Paris, France
- Centre de Recherche Saint Antoine, INSERM UMRS_938 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| |
Collapse
|
21
|
Ferrari C, Ashraf B, Saeed Z, Tadros M. Understanding Why Metabolic-Dysfunction-Associated Steatohepatitis Lags Behind Hepatitis C in Therapeutic Development and Treatment Advances. GASTROENTEROLOGY INSIGHTS 2024; 15:944-962. [DOI: 10.3390/gastroent15040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
Therapeutic development for metabolic-dysfunction-associated steatohepatitis (MASH) trails behind the success seen in hepatitis C virus (HCV) management. HCV, characterized by a viral etiology, benefits from direct-acting antivirals (DAAs) targeting viral proteins, achieving cure rates exceeding 90%. In contrast, MASH involves complex metabolic, genetic, and environmental factors, presenting challenges for drug development. Non-invasive diagnostics like ultrasound, FibroScan, and serum biomarkers, while increasingly used, lack the diagnostic accuracy of liver biopsy, the current gold standard. This review evaluates therapies for MASH, including resmetirom (Rezdiffra) and combinations like pioglitazone and vitamin E, which show potential but offer modest improvements due to MASH’s heterogeneity. The limited efficacy of these treatments highlights the need for multi-targeted strategies addressing metabolic and fibrotic components. Drawing parallels to HCV’s success, this review emphasizes advancing diagnostics and therapies for MASH. Developing effective, patient-specific therapies is crucial to closing the gap between MASH and better-managed liver diseases, optimizing care for this growing health challenge.
Collapse
Affiliation(s)
- Caesar Ferrari
- Department of Gastroenterology and Hepatology, Albany Medical College, Albany, NY 12208, USA
| | - Bilal Ashraf
- HCA Houston Healthcare Kingwood, Kingwood, TX 77339, USA
| | - Zainab Saeed
- Houston Methodist Baytown Hospital, Baytown, TX 77521, USA
| | - Micheal Tadros
- Department of Gastroenterology and Hepatology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
22
|
Zhang YF, Qiao W, Zhuang J, Feng H, Zhang Z, Zhang Y. Association of ultra-processed food intake with severe non-alcoholic fatty liver disease: a prospective study of 143073 UK Biobank participants. J Nutr Health Aging 2024; 28:100352. [PMID: 39340900 DOI: 10.1016/j.jnha.2024.100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Previous studies indicate a link between non-alcoholic fatty liver disease (NAFLD) and unhealthy dietary patterns or nutrient intake. However, it remains unclear whether ultra-processed foods (UPF) contribute to an increased risk of NAFLD. This study aimed to explore how ultra-processed food consumption correlates with severe NAFLD using the UK Biobank data. METHODS This prospective cohort study included 143,073 participants from the UK Biobank. UPF consumption levels were determined using the NOVA classification and quantified from 24-h dietary recall data. The association between UPF consumption and severe NAFLD (hospitalization or death) was initially examined using Cox proportional hazards models with intake quartiles. Nonlinear associations were investigated using penalized cubic splines fitted in the Cox proportional hazards models. Adjustments were made for general characteristics, sociodemographic factors, body mass index (BMI), and lifestyle. RESULTS Throughout the median follow-up period of 10.5 years, 1,445 participants developed severe NAFLD. The adjusted models indicated a significant increase in severe NAFLD risk in higher UPF intake groups compared to the lowest quartile (HR: 1.26 [95% CI: 1.11-1.43]). Subgroup analysis revealed that individuals with a BMI of 25 or higher were at greater risk in the highest quartile of UPF consumption. Sensitivity analyses yielded results consistent with these findings. CONCLUSION Higher consumption of UPF is associated with an increased risk of severe NAFLD. Reducing the intake of UPF can be a potential approach to lower the risk of NAFLD.
Collapse
Affiliation(s)
- Yi-Feng Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wanning Qiao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jinhong Zhuang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Hanxiao Feng
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhilan Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Zhang M, Guo C, Li Z, Cai X, Wen X, Lv F, Lin C, Ji L. Mulberry Twig Alkaloids Improved the Progression of Metabolic-Associated Fatty Liver Disease in High-Fat Diet-Induced Obese Mice by Regulating the PGC1α/PPARα and KEAP1/NRF2 Pathways. Pharmaceuticals (Basel) 2024; 17:1287. [PMID: 39458927 PMCID: PMC11514595 DOI: 10.3390/ph17101287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Metabolic-associated fatty liver disease (MAFLD) is one of the most common liver disorders associated with obesity and metabolic syndrome, and poses a significant global health burden with limited effective treatments. The aim of this study was to assess the protective effects of mulberry twig alkaloids (SZ-A) on MAFLD and to further investigate the underlying mechanisms including the specific targets or pathways. Methods: Diet-induced obesity (DIO) and normal mouse models were established by feeding C57Bl/6J mice with a high-fat diet (HFD) or common diet for 12 weeks. SZ-A, dapagliflozin, and placebo were administered to corresponding mouse groups for 8 weeks. Data of fasting blood glucose, glucose tolerance, insulin tolerance, and the body weight of mice were collected at the baseline and termination of the experiment. Serum liver enzymes and lipids were measured by ELISA. Western blotting, qPCR, and pathological section staining were implemented to evaluate the degrees of liver steatosis, fibrosis, and oxidative stress in mice. Results: In DIO mouse models, high-dose SZ-A (800 mg/kg/d) treatment significantly inhibited HFD-induced weight gain, improved insulin tolerance, and reduced serum alanine aminotransferase, total cholesterol, and triglyceride levels compared with placebo. In DIO mice, SZ-A could alleviate the pathological changes of hepatic steatosis and fibrosis compared with placebo. Lipid catabolism and antioxidant stress-related proteins were significantly increased in the livers of the high-dose SZ-A group (p < 0.05). Inhibition of PGC1α could inhibit the function of SZ-A to enhance lipid metabolism in hepatocytes. PGC1α might interact with NRF2 to exert MAFLD-remedying effects. Conclusions: By regulating the expression of PGC1α and its interacting KEAP1/NRF2 pathway in mouse liver cells, SZ-A played important roles in regulating lipid metabolism, inhibiting oxidative stress, and postponing liver fibrosis in mice with MAFLD.
Collapse
Affiliation(s)
| | | | | | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing 100044, China
| | | | | | | | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
24
|
Bott S, Lallement J, Marino A, Daskalopoulos EP, Beauloye C, Esfahani H, Dessy C, Leclercq IA. When the liver is in poor condition, so is the heart - cardiac remodelling in MASH mouse models. Clin Sci (Lond) 2024; 138:1151-1171. [PMID: 39206703 PMCID: PMC11405860 DOI: 10.1042/cs20240833] [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: 05/02/2024] [Revised: 08/15/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) confers a risk for cardiovascular diseases in patients. Animal models may help exploring the mechanisms linking liver and heart diseases. Hence, we explored the cardiac phenotype in two MASH mouse models: foz/foz mice fed a high-fat diet (HFD) for 24 or 60 weeks and C57BL/6J mice fed a high-fat-, high-cholesterol-, and high-fructose diet for 60 weeks. Angiotensin II (AngII) was used as an additional cardiovascular stressor for 4 weeks in 10 weeks HFD-fed foz/foz mice. Foz/foz mice with fibrosing MASH developed cardiac hypertrophy with adverse cardiac remodelling not seen in WT similarly fed the HFD. AngII caused hypertension and up-regulated the expression of genes contributing to pathological cardiac hypertrophy (Nppa, Myh7) more severely so in foz/foz mice than in controls. After 60 weeks of HFD, while liver disease had progressed to burn-out non steatotic MASH with hepatocellular carcinoma in 50% of the animals, the cardiomyopathy did not. In an independent model (C57BL/6J mice fed a fat-, cholesterol- and fructose-rich diet), moderate fibrosing MASH is associated with cardiac fibrosis and dysregulation of genes involved in pathological remodelling (Col1a1, Col3a1, Vim, Myh6, Slc2a1). Thus, animals with MASH present consistent adverse structural changes in the heart with no patent alteration of cardiac function even when stressed with exogenous AngII. Liver disease, and likely not overfeeding or aging alone, is associated with this cardiac phenotype. Our findings support foz/foz mice as suitable for studying links between MASH and heart structural changes ahead of heart failure.
Collapse
Affiliation(s)
- Sebastian Bott
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Justine Lallement
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Alice Marino
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | | | - Christophe Beauloye
- Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
- Division of Cardiology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Hrag Esfahani
- Platform of Integrated Physiology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle Anne Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels
| |
Collapse
|
25
|
Nardin GD, Colombo BDS, Ronsoni MF, Silva PESE, Fayad L, Wildner LM, Bazzo ML, Dantas-Correa EB, Narciso-Schiavon JL, Schiavon LDL. Thyroid hormone profile is related to prognosis in acute decompensation of cirrhosis. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230249. [PMID: 39420934 PMCID: PMC11460973 DOI: 10.20945/2359-4292-2023-0249] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 11/21/2023] [Indexed: 10/19/2024]
Abstract
OBJECTIVE To investigate the prognostic significance of thyroid hormone profile in patients hospitalized for decompensated cirrhosis. SUBJECTS AND METHODS Prospective cohort study that included 119 subjects. All subjects were evaluated at admission and followed for 90 days. TSH, fT3, fT4 were measured within 24 hours of hospitalization. RESULTS Higher fT4 and lower fT3 levels were observed among Child-Pugh C patients as compared to Child-Pugh A and B, and in those with acute-on-chronic liver failure (ACLF). Lower fT3/fT4 ratio was observed in those with ascites, infections, ACLF, and in Child-Pugh C. Ninety-day mortality was 26.9% and it was independently associated with higher Model for End-stage Liver Disease (MELD) and TSH, and lower fT3/fT4 ratio in multivariate analysis. A new prognostic model including MELD, TSH and fT3/fT4 ratio was devised. The areas under the receiver operating characteristic curves for MELD, fT3/fT4 ratio, TSH (μIU/mL), and the new model for predicting 90-day mortality were 0.847 ± 0.041, 0.841 ± 0.039, 0.658 ± 0.062, and 0.899 ± 0.031, respectively. The 90-day survival was 31.6% in patients with values of the predictive model ≥ -0.77 and 93.5% for values < -0.77 (P < 0.001). CONCLUSIONS Thyroid hormone profile was strongly associated with worse outcomes in patients with cirrhosis and might represent promising prognostic tools that can be incorporated in clinical practice.
Collapse
Affiliation(s)
- Giovana De Nardin
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Bruno da Silveira Colombo
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Marcelo Fernando Ronsoni
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Pedro Eduardo Soares e Silva
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Leonardo Fayad
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Letícia Muraro Wildner
- Departamento de Análises ClínicasUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Departamento de Análises Clínicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Maria Luiza Bazzo
- Departamento de Análises ClínicasUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Departamento de Análises Clínicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Esther Buzaglo Dantas-Correa
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Janaína Luz Narciso-Schiavon
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| | - Leonardo de Lucca Schiavon
- Divisão de GastroenterologiaUniversidade Federal de Santa CatarinaFlorianópolisSCBrasil Divisão de Gastroenterologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| |
Collapse
|
26
|
Chen K, Wei L, Yu S, He N, Zhang F. Identification of autophagy-related signatures in nonalcoholic fatty liver disease and correlation with non-parenchymal cells of the liver. Mol Omics 2024; 20:469-482. [PMID: 38982979 DOI: 10.1039/d4mo00060a] [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: 07/11/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic hepatic disease. The incidence and prevalence of NAFLD have increased greatly in recent years, and there is still a lack of effective drugs. Autophagy plays an important role in promoting liver metabolism and maintaining liver homeostasis, and defects in autophagy levels are considered to be related to the development of NAFLD. However, the molecular mechanisms of autophagy in NAFLD still remain unknown. In this study, we identified 6 autophagy-associated hub genes using gene expression profiles obtained from the GSE48452 and GSE89632 datasets. Biomarkers were screened according to gene significance (GS) and module membership (MM) using weighted gene co-expression network analysis (WGCNA), and the immune infiltration landscape of the liver in NAFLD patients was explored using the CIBERSORT algorithm. Subsequently, we analyzed the relationship between liver non-parenchymal cells and autophagy-related hub genes using scRNA-seq data (GSE129516). Finally, we separated the NAFLD patients into two groups based on 6 hub genes by consensus clustering and screened 10 potential autophagy-related small molecules based on the cMAP database.
Collapse
Affiliation(s)
- Kaiwei Chen
- Department of Infectious Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266003, China.
| | - Ling Wei
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| | - Shengnan Yu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266003, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266003, China.
| | - Fengjuan Zhang
- Department of Infectious Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
27
|
Zhu H, Zhao T, Zhao S, Yang S, Jiang K, Li S, Kang Y, Yang Z, Shen J, Shen S, Tao H, Xuan J, Yang M, Xu B, Wang F, Jiang M. O-GlcNAcylation promotes the progression of nonalcoholic fatty liver disease by upregulating the expression and function of CD36. Metabolism 2024; 156:155914. [PMID: 38642829 DOI: 10.1016/j.metabol.2024.155914] [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: 01/17/2024] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD) and its progressive variant, nonalcoholic steatohepatitis (NASH), constitute a burgeoning worldwide epidemic with no FDA-approved pharmacotherapies. The multifunctional immunometabolic receptor, fatty acid translocase CD36 (CD36), plays an important role in the progression of hepatic steatosis. O-GlcNAcylation is a crucial posttranslational modification that mediates the distribution and function of CD36, but its involvement in NAFLD remains poorly understood. METHODS O-GlcNAcylation and CD36 expression were evaluated in human liver tissues obtained from NASH patients and normal control. Mice with hepatocyte-specific CD36 knockout were administered adeno-associated viral vectors expressing wild-type CD36 (WT-CD36) or CD36 O-GlcNAcylation site mutants (S468A&T470A-CD36) and were provided with a high-fat/high-cholesterol (HFHC) diet for 3 months. RT-qPCR analysis, immunoblotting, dual-luciferase reporter assays, chromatin immunoprecipitation, and coimmunoprecipitation were performed to explore the mechanisms by which O-GlcNAcylation regulates CD36 expression. Membrane protein extraction, immunofluorescence analysis, site-directed mutagenesis, and fatty acid uptake assays were conducted to elucidate the impact of O-GlcNAcylation on CD36 function. RESULTS O-GlcNAcylation and CD36 expression were significantly increased in patients with NASH, mouse models of NASH, and palmitic acid-stimulated hepatocytes. Mechanistically, the increase in O-GlcNAcylation facilitated the transcription of CD36 via the NF-κB signalling pathway and stabilized the CD36 protein by inhibiting its ubiquitination, thereby promoting CD36 expression. On the other hand, O-GlcNAcylation facilitated the membrane localization of CD36, fatty acid uptake, and lipid accumulation. However, site-directed mutagenesis of residues S468 and T470 of CD36 reversed these effects. Furthermore, compared with their WT-CD36 counterparts, HFHC-fed S468A&T470A-CD36 mice exhibited decreases in systemic insulin resistance, steatosis severity, inflammation and fibrosis. Pharmacological inhibition of O-GlcNAcylation and CD36 also mitigated the progression of NASH. CONCLUSIONS O-GlcNAcylation promotes the progression of NAFLD by upregulating CD36 expression and function. Inhibition of CD36 O-GlcNAcylation protects against NASH, highlighting a potentially effective therapeutic approach for individuals with NASH.
Collapse
Affiliation(s)
- Hanlong Zhu
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Tianming Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China.
| | - Si Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Suzhen Yang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Kang Jiang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Shupei Li
- Department of Gastroenterology, Nanjing University of Chinese Medicine, Jinling School of Clinical Medicine, Nanjing, Jiangsu, China.
| | - Ying Kang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Zhuoxin Yang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Jiajia Shen
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Si Shen
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Hui Tao
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Ji Xuan
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Miaofang Yang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Bing Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Fangyu Wang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Mingzuo Jiang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Song DS, Kim HY, Jung YK, Kim TH, Yim HJ, Yoon EL, Suk KT, Yoo JJ, Kim SG, Kim MY, Chang Y, Jeong SW, Jang JY, Kim SE, Kim JH, Park JG, Kim W, Yang JM, Kim DJ, Choudhury AK, Arora V, Sarin SK. Dynamic analysis of acute deterioration in chronic liver disease patients using modified quick sequential organ failure assessment. Clin Mol Hepatol 2024; 30:388-405. [PMID: 38600873 PMCID: PMC11261223 DOI: 10.3350/cmh.2023.0563] [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: 12/27/2023] [Revised: 03/23/2024] [Accepted: 04/09/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND/AIMS Quick sequential organ failure assessment (qSOFA) is believed to identify patients at risk of poor outcomes in those with suspected infection. We aimed to evaluate the ability of modified qSOFA (m-qSOFA) to identify high-risk patients among those with acutely deteriorated chronic liver disease (CLD), especially those with acute-onchronic liver failure (ACLF). METHODS We used data from both the Korean Acute-on-Chronic Liver Failure (KACLiF) and the Asian Pacific Association for the Study of the Liver ACLF Research Consortium (AARC) cohorts. qSOFA was modified by replacing the Glasgow Coma Scale with hepatic encephalopathy, and an m-qSOFA ≥2 was considered high. RESULTS Patients with high m-qSOFA had a significantly lower 1-month transplant-free survival (TFS) in both cohorts and higher organ failure development in KACLiF than those with low m-qSOFA (Ps<0.05). Subgroup analysis by ACLF showed that patients with high m-qSOFA had lower TFS than those with low m-qSOFA. m-qSOFA was an independent prognostic factor (hazard ratios, HR=2.604, 95% confidence interval, CI 1.353-5.013, P=0.004 in KACLiF and HR=1.904, 95% CI 1.484- 2.442, P<0.001 in AARC). The patients with low m-qSOFA at baseline but high m-qSOFA on day 7 had a significantly lower 1-month TFS than those with high m-qSOFA at baseline but low m-qSOFA on day 7 (52.6% vs. 89.4%, P<0.001 in KACLiF and 26.9% vs. 61.5%, P<0.001 in AARC). CONCLUSION Baseline and dynamic changes in m-qSOFA may identify patients with a high risk of developing organ failure and short-term mortality among CLD patients with acute deterioration.
Collapse
Affiliation(s)
- Do Seon Song
- Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Hee Yeon Kim
- Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Young Kul Jung
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Tae Hyung Kim
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Hyung Joon Yim
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Eileen L Yoon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Ki Tae Suk
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Jeong-ju Yoo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sang Gyune Kim
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Chang
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Soung Won Jeong
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Jae Young Jang
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Sung-Eun Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Jung-Hee Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Jung Gil Park
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Jin Mo Yang
- Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Dong Joon Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Korean Acute-on-Chronic Liver Failure (KACLiF) study group
- Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | | | - Vinod Arora
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - APASL ACLF Research Consortium (AARC) for APASL ACLF working party
- Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
29
|
Navarro-Masip È, Mestres Petit N, Salinas-Roca B, Herrerías F, Vilardell F, de la Fuente MC, Pallares J, Santamaría M, Zorzano-Martínez M, Sánchez E, Matías-Guiu X, López-Cano C, Soler AG, León-Mengíbar J, Bueno M, Lecube A. Metabolic Dysfunction-Associated Steatotic Liver Disease in Severe Obesity and Concordance between Invasive (Biopsy) and Noninvasive (OWLiver®) Diagnoses. Obes Facts 2024; 17:473-482. [PMID: 38934179 PMCID: PMC11540414 DOI: 10.1159/000538765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/05/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD), now termed metabolic dysfunction-associated steatotic liver disease (MASLD), is an escalating health concern linked to obesity and type 2 diabetes. Despite liver biopsy being the gold standard, its invasiveness underscores the need for noninvasive diagnostic methods. METHODS A cross-sectional study was performed to assess MASLD using the noninvasive OWLiver® serum lipidomics test in a cohort of 117 patients with severe obesity undergoing bariatric surgery, comparing outcomes with liver biopsy. Exclusions (n = 24) included insufficient data, liver disease etiology other than MASLD, corticosteroid treatment, excessive alcohol consumption, low glomerular filtration rate, and declination to participate. Comprehensive laboratory tests, demographic assessments, and liver biopsies were performed. Serum metabolites were analyzed using OWLiver®, a serum lipidomic test that discriminates between healthy liver, steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and MASH with fibrosis ≥2 by means of three algorithms run sequentially. RESULTS Liver biopsy revealed a MASLD prevalence of 95.7%, with MASH present in 28.2% of cases. OWLiver® demonstrated a tendency to diagnose more severe cases. Body mass index (BMI), rather than the presence of type 2 diabetes, emerged as the sole independent factor linked to the probability of concordance. Therefore, the all-population concordance of 63.2% between OWLiver® and liver biopsy notably raised to 77.1% in patients with a BMI <40 kg/m2. These findings suggest a potential correlation between lower BMI and enhanced concordance between OWLiver® and biopsy. CONCLUSION This study yields valuable insights into the concordance between liver biopsy and the noninvasive serum lipidomic test, OWLiver®, in severe obesity. OWLiver® demonstrated a tendency to amplify MASLD severity, with BMI values influencing concordance. Patients with BMI <40 kg/m2 may derive optimal benefits from this noninvasive diagnostic approach.
Collapse
Affiliation(s)
- Èlia Navarro-Masip
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain,
| | - Nuria Mestres Petit
- General and Digestive Surgery Department, Arnau de Vilanova University Hospital, Lleida, Spain
- Surgery Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Blanca Salinas-Roca
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Facultat de Ciències de la Salut-Universitat Ramón Llull Blanquerna, Carrer Padilla, Barcelona, Spain
| | - Fernando Herrerías
- General and Digestive Surgery Department, Arnau de Vilanova University Hospital, Lleida, Spain
- Surgery Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Felip Vilardell
- Department of Pathology and Molecular Genetics, Arnau de Vilanova University Hospital, Institut de Recerca Biomèdica (IRB), Lleida, Spain
- Oncological Pathology Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Mari Cruz de la Fuente
- General and Digestive Surgery Department, Arnau de Vilanova University Hospital, Lleida, Spain
- Surgery Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Judit Pallares
- Department of Pathology and Molecular Genetics, Arnau de Vilanova University Hospital, Institut de Recerca Biomèdica (IRB), Lleida, Spain
- Oncological Pathology Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Maite Santamaría
- General and Digestive Surgery Department, Arnau de Vilanova University Hospital, Lleida, Spain
- Surgery Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Marta Zorzano-Martínez
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Enric Sánchez
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Xavier Matías-Guiu
- Department of Pathology and Molecular Genetics, Arnau de Vilanova University Hospital, Institut de Recerca Biomèdica (IRB), Lleida, Spain
- Oncological Pathology Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Carolina López-Cano
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Ana Gloria Soler
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Josep León-Mengíbar
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Marta Bueno
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, Lleida, Spain
| | - Albert Lecube
- Obesity, Diabetes and Metabolism (ODIM) Research Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
- Endocrinology and Nutrition Department, Arnau de Vilanova University Hospital, Lleida, Spain
| |
Collapse
|
30
|
Verhoeven A, van Ertvelde J, Boeckmans J, Gatzios A, Jover R, Lindeman B, Lopez-Soop G, Rodrigues RM, Rapisarda A, Sanz-Serrano J, Stinckens M, Sepehri S, Teunis M, Vinken M, Jiang J, Vanhaecke T. A quantitative weight-of-evidence method for confidence assessment of adverse outcome pathway networks: A case study on chemical-induced liver steatosis. Toxicology 2024; 505:153814. [PMID: 38677583 DOI: 10.1016/j.tox.2024.153814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024]
Abstract
The field of chemical toxicity testing is undergoing a transition to overcome the limitations of in vivo experiments. This evolution involves implementing innovative non-animal approaches to improve predictability and provide a more precise understanding of toxicity mechanisms. Adverse outcome pathway (AOP) networks are pivotal in organizing existing mechanistic knowledge related to toxicological processes. However, these AOP networks are dynamic and require regular updates to incorporate the latest data. Regulatory challenges also persist due to concerns about the reliability of the information they offer. This study introduces a generic Weight-of-Evidence (WoE) scoring method, aligned with the tailored Bradford-Hill criteria, to quantitatively assess the confidence levels in key event relationships (KERs) within AOP networks. We use the previously published AOP network on chemical-induced liver steatosis, a prevalent form of human liver injury, as a case study. Initially, the existing AOP network is optimized with the latest scientific information extracted from PubMed using the free SysRev platform for artificial intelligence (AI)-based abstract inclusion and standardized data collection. The resulting optimized AOP network, constructed using Cytoscape, visually represents confidence levels through node size (key event, KE) and edge thickness (KERs). Additionally, a Shiny application is developed to facilitate user interaction with the dataset, promoting future updates. Our analysis of 173 research papers yielded 100 unique KEs and 221 KERs among which 72 KEs and 170 KERs, respectively, have not been previously documented in the prior AOP network or AOP-wiki. Notably, modifications in de novo lipogenesis, fatty acid uptake and mitochondrial beta-oxidation, leading to lipid accumulation and liver steatosis, garnered the highest KER confidence scores. In conclusion, our study delivers a generic methodology for developing and assessing AOP networks. The quantitative WoE scoring method facilitates in determining the level of support for KERs within the optimized AOP network, offering valuable insights into its utility in both scientific research and regulatory contexts. KERs supported by robust evidence represent promising candidates for inclusion in an in vitro test battery for reliably predicting chemical-induced liver steatosis within regulatory frameworks.
Collapse
Affiliation(s)
- Anouk Verhoeven
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jonas van Ertvelde
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joost Boeckmans
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alexandra Gatzios
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ramiro Jover
- Joint Research Unit in Experimental Hepatology, University of Valencia, Health Research Institute Hospital La Fe & CIBER of Hepatic and Digestive Diseases, Valencia, Spain
| | - Birgitte Lindeman
- Department of Chemical Toxicology, Norwegian Institute of Public Health, Oslo, Norway
| | - Graciela Lopez-Soop
- Department of Chemical Toxicology, Norwegian Institute of Public Health, Oslo, Norway
| | - Robim M Rodrigues
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Anna Rapisarda
- Joint Research Unit in Experimental Hepatology, University of Valencia, Health Research Institute Hospital La Fe & CIBER of Hepatic and Digestive Diseases, Valencia, Spain
| | - Julen Sanz-Serrano
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marth Stinckens
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sara Sepehri
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marc Teunis
- Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences Utrecht, Utrecht, the Netherlands
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jian Jiang
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
31
|
Buss C, Marette A, Escouto GS, Pilon G, Tovo CV. Reply to F Du and L Tao. J Nutr 2024; 154:1960. [PMID: 38599386 DOI: 10.1016/j.tjnut.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Affiliation(s)
- Caroline Buss
- Graduate Study Program (GSP) in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre-RS, Brazil; Graduate Study Program in Hepatology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre-RS, Brazil; Nutrition Department, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre-RS, Brazil.
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Canada; Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
| | - Giselle S Escouto
- Graduate Study Program (GSP) in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre-RS, Brazil
| | - Geneviève Pilon
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Canada; Institute of Nutrition and Functional Foods (INAF), Laval University, Québec, Canada
| | - Cristiane V Tovo
- Graduate Study Program in Hepatology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre-RS, Brazil
| |
Collapse
|
32
|
Iwaki M, Yoneda M, Wada N, Otani T, Kobayashi T, Nogami A, Saito S, Nakajima A. Emerging drugs for the treatment of hepatic fibrosis on nonalcoholic steatohepatitis. Expert Opin Emerg Drugs 2024; 29:127-137. [PMID: 38469871 DOI: 10.1080/14728214.2024.2328036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Approved drug therapies for nonalcoholic steatohepatitis (NASH) are lacking, for which various agents are currently being tested in clinical trials. Effective drugs for liver fibrosis, the factor most associated with prognosis in NASH, are important. AREAS COVERED This study reviewed the treatment of NASH with a focus on the effects of existing drugs and new drugs on liver fibrosis. EXPERT OPINION Considering the complex pathophysiology of fibrosis in NASH, drug therapy may target multiple pathways. The method of assessing fibrosis is important when considering treatment for liver fibrosis in NASH. The Food and Drug Administration considers an important fibrosis endpoint to be histological improvement in at least one fibrosis stage while preventing worsening of fatty hepatitis. To obtain approval as a drug for NASH, efficacy needs to be demonstrated on endpoints such as liver-related events and myocardial infarction. Among the current therapeutic agents for NASH, thiazolidinedione, sodium-glucose co-transporter 2, and selective peroxisome proliferator-activated receptors α modulator have been reported to be effective against fibrosis, although further evidence is required. The effects of pan-peroxisome proliferator-activated receptors, obeticholic acid, and fibroblast growth factor-21 analogs on liver fibrosis in the development stage therapeutics for NASH are of particular interest.
Collapse
Affiliation(s)
- Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Naohiro Wada
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomohiro Otani
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoru Saito
- Department of Gastroenterology, Sanno Hospital, Minato-Ku, Tokyo, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
33
|
Tincopa MA, Anstee QM, Loomba R. New and emerging treatments for metabolic dysfunction-associated steatohepatitis. Cell Metab 2024; 36:912-926. [PMID: 38608696 DOI: 10.1016/j.cmet.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a leading etiology of chronic liver disease worldwide, with increasing incidence and prevalence in the setting of the obesity epidemic. MASH is also a leading indication for liver transplantation, given its associated risk of progression to end-stage liver disease. A key challenge in managing MASH is the lack of approved pharmacotherapy. In its absence, lifestyle interventions with a focus on healthy nutrition and regular physical activity have been the cornerstone of therapy. Real-world efficacy and sustainability of lifestyle interventions are low, however. Pharmacotherapy development for MASH is emerging with promising data from several agents with different mechanisms of action (MOAs) in phase 3 clinical trials. In this review, we highlight ongoing challenges and potential solutions in drug development for MASH and provide an overview of available data from emerging therapies across multiple MOAs.
Collapse
Affiliation(s)
- Monica A Tincopa
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA 92103, USA
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Newcastle NIHR Biomedical Research Center, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, University of California, San Diego, La Jolla, CA 92103, USA; School of Public Health, University of California, San Diego, La Jolla, CA 92103, USA.
| |
Collapse
|
34
|
Fischer AM, Lechea N, Coxson HO. This Is What Metabolic Dysfunction-Associated Steatotic Liver Disease Looks Like: Potential of a Multiparametric MRI Protocol. Semin Liver Dis 2024; 44:226-238. [PMID: 38806158 DOI: 10.1055/a-2334-8525] [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: 05/30/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent condition with a broad spectrum defined by liver biopsy. This gold standard method evaluates three features: steatosis, activity (ballooning and lobular inflammation), and fibrosis, attributing them to certain grades or stages using a semiquantitative scoring system. However, liver biopsy is subject to numerous restrictions, creating an unmet need for a reliable and reproducible method for MASLD assessment, grading, and staging. Noninvasive imaging modalities, such as magnetic resonance imaging (MRI), offer the potential to assess quantitative liver parameters. This review aims to provide an overview of the available MRI techniques for the three criteria evaluated individually by liver histology. Here, we discuss the possibility of combining multiple MRI parameters to replace liver biopsy with a holistic, multiparametric MRI protocol. In conclusion, the development and implementation of such an approach could significantly improve the diagnosis and management of MASLD, reducing the need for invasive procedures and paving the way for more personalized treatment strategies.
Collapse
Affiliation(s)
- Anja M Fischer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nazim Lechea
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Harvey O Coxson
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
35
|
Helgesson S, Tarai S, Langner T, Ahlström H, Johansson L, Kullberg J, Lundström E. Spleen volume is independently associated with non-alcoholic fatty liver disease, liver volume and liver fibrosis. Heliyon 2024; 10:e28123. [PMID: 38665588 PMCID: PMC11043861 DOI: 10.1016/j.heliyon.2024.e28123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) can lead to irreversible liver damage manifesting in systemic effects (e.g., elevated portal vein pressure and splenomegaly) with increased risk of deadly outcomes. However, the association of spleen volume with NAFLD and related type 2-diabetes (T2D) is not fully understood. The UK Biobank contains comprehensive health-data of 500,000 participants, including clinical data and MR images of >40,000 individuals. The present study estimated the spleen volume of 37,066 participants through automated deep learning-based image segmentation of neck-to-knee MR images. The aim was to investigate the associations of spleen volume with NAFLD, T2D and liver fibrosis, while adjusting for natural confounders. The recent redefinition and new designation of NAFLD to metabolic dysfunction-associated steatotic liver disease (MASLD), promoted by major organisations of studies on liver disease, was not employed as introduced after the conduct of this study. The results showed that spleen volume decreased with age, correlated positively with body size and was smaller in females compared to males. Larger spleens were observed in subjects with NAFLD and T2D compared to controls. Spleen volume was also positively and independently associated with liver fat fraction, liver volume and the fibrosis-4 score, with notable volumetric increases already at low liver fat fractions and volumes, but not independently associated with T2D. These results suggest a link between spleen volume and NAFLD already at an early stage of the disease, potentially due to initial rise in portal vein pressure.
Collapse
Affiliation(s)
- Samuel Helgesson
- Radiology, Department of Surgical Sciences, Uppsala University, Sweden
| | - Sambit Tarai
- Radiology, Department of Surgical Sciences, Uppsala University, Sweden
- Antaros Medical AB, BioVenture Hub, Sweden
| | | | - Håkan Ahlström
- Radiology, Department of Surgical Sciences, Uppsala University, Sweden
- Antaros Medical AB, BioVenture Hub, Sweden
| | | | - Joel Kullberg
- Radiology, Department of Surgical Sciences, Uppsala University, Sweden
- Antaros Medical AB, BioVenture Hub, Sweden
| | - Elin Lundström
- Radiology, Department of Surgical Sciences, Uppsala University, Sweden
| |
Collapse
|
36
|
Ciardullo S, Muraca E, Vergani M, Invernizzi P, Perseghin G. Advancements in pharmacological treatment of NAFLD/MASLD: a focus on metabolic and liver-targeted interventions. Gastroenterol Rep (Oxf) 2024; 12:goae029. [PMID: 38681750 PMCID: PMC11052658 DOI: 10.1093/gastro/goae029] [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: 02/13/2024] [Accepted: 02/27/2024] [Indexed: 05/01/2024] Open
Abstract
In the present narrative review, we have summarized evidence on the pharmacological treatment of non-alcoholic fatty liver disease (NAFLD)/metabolic dysfunction-associated steatotic liver disease (MASLD). We start by reviewing the epidemiology of the condition and its close association with obesity and type 2 diabetes. We then discuss how randomized-controlled trials are performed following guidance from regulatory agencies, including differences and similarities between requirements of the US Food and Drug Administration and the European Medicine Agency. Difficulties and hurdles related to limitations of liver biopsy, a large number of screening failures in recruiting patients, as well as unpredictable response rates in the placebo group are evaluated. Finally, we recapitulate the strategies employed for potential drug treatments of this orphan condition. The first is to repurpose drugs that originally targeted T2DM and/or obesity, such as pioglitazone, glucagon-like peptide 1 receptor agonists (liraglutide and semaglutide), multi-agonists (tirzepatide and retatrutide), and sodium-glucose transporter 2 inhibitors. The second is to develop drugs specifically targeting NAFLD/MASLD. Among those, we focused on resmetirom, fibroblast growth factor 21 analogs, and lanifibranor, as they are currently in Phase 3 of their clinical trial development. While many failures have characterized the field of pharmacological treatment of NAFLD/MASLD in the past, it is likely that approval of the first treatments is near. As occurs in many chronic conditions, combination therapy might lead to better outcomes. In the case of non-alcoholic steatohepatitis, we speculate that drugs treating underlying metabolic co-morbidities might play a bigger role in the earlier stages of disease, while liver-targeting molecules will become vital in patients with more advanced disease in terms of inflammation and fibrosis.
Collapse
Affiliation(s)
- Stefano Ciardullo
- Department of Medicine and Rehabilitation, Policlinico di Monza, Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Emanuele Muraca
- Department of Medicine and Rehabilitation, Policlinico di Monza, Monza, Italy
| | - Michela Vergani
- Department of Medicine and Rehabilitation, Policlinico di Monza, Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER) San Gerardo Hospital, Monza, Italy
| | - Gianluca Perseghin
- Department of Medicine and Rehabilitation, Policlinico di Monza, Monza, Italy
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| |
Collapse
|
37
|
Chen H, Shen H, Han J, Wang P, Song D, Shen H, Wei X, Yang B, Li J. Performance of ATT and UDFF in the diagnosis of non-alcoholic fatty liver: An animal experiment. Heliyon 2024; 10:e27993. [PMID: 38560108 PMCID: PMC10981026 DOI: 10.1016/j.heliyon.2024.e27993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/01/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
Objective To establish a Bama minipigs model with Non-Alcoholic Fatty Liver (NAFL) induced by a high-fat diet and investigate the application of attenuation coefficient (ATT) and ultrasound-derived fat fraction (UDFF) in the diagnosis of NAFL. Methods Six-month-old male Bama minipigs were randomly divided into normal control and high-fat groups (n = 3 pigs per group), and fed with a control diet and high-fat diet for 32 weeks. Weight and body length were measured every four weeks, followed by quantitative ultrasound imaging (ATT and UDFF), blood biochemical markers, and liver biopsies on the same day. Using the Non-Alcoholic Fatty Liver Disease (NAFLD) Activity Score (NAS) as a reference, we analyzed the correlation between ATT, UDFF, and their score results. Results Compared with the normal control group, the body weight, body mass index (BMI), and serum levels of triglyceride (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) in the High-fat group were significantly different at Week 12 (P < 0.05). Spearman correlation analysis showed that the ATT value was significantly correlated with NAS score (r = 0.76, P < 0.001), and the UDFF value was significantly correlated with NAS score (r = 0.80, P < 0.001). The optimal cut-off value of ATT and UDFF were 0.59 dB/cm/MHz and 5.5%, respectively. These values are optimal for diagnosis of NAFL in Bama minipig model. Conclusion ATT and UDFF have a high correlation with steatosis, and can be used as a non-invasive method for early screening of hepatic steatosis, which can dynamically monitor the change of disease course.
Collapse
Affiliation(s)
- Huihui Chen
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Huiming Shen
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jiahao Han
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Pingping Wang
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Danlei Song
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Hongyuan Shen
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xiaoying Wei
- Department of Pathology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Bingjie Yang
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jia Li
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| |
Collapse
|
38
|
Allen AM, Charlton M, Cusi K, Harrison SA, Kowdley KV, Noureddin M, Shubrook JH. Guideline-based management of metabolic dysfunction-associated steatotic liver disease in the primary care setting. Postgrad Med 2024; 136:229-245. [PMID: 38465573 DOI: 10.1080/00325481.2024.2325332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/26/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing worldwide. Primary care providers play a critical role in the screening, diagnosis, and management of MASLD and/or metabolic dysfunction-associated steatohepatitis (MASH), though they can face challenges in this setting, particularly where healthcare resources are limited and barriers to care exist. To address these challenges, several guidelines have been developed to provide evidence-based recommendations for the clinical assessment and management of patients with MASLD/MASH. AIMS To provide a unified, simple-to-understand, practical guide for MASLD screening, diagnosis, and management based on current guideline recommendations, for use by primary care providers in daily practice. METHODS Evidence-based recommendations from several international guidelines were summarized, focusing on the similarities and differences between them. RESULTS Recommendations are broadly aligned across the guidelines, but several key differences are evident. Practical guidance is provided on screening, identifying target populations for risk stratification, initial evaluation of individuals with suspected MASLD, surveillance, risk stratification and referral, as well as approaches to the management of MASLD and associated comorbidities, with specific considerations for the primary care setting. CONCLUSIONS Primary care providers are ideally placed to identify at-risk individuals, implement evidence-based interventions to prevent the development of fibrosis and cirrhosis, and effectively manage comorbidities. Equipping primary care providers with the necessary knowledge and tools to effectively manage MASLD/MASH may help to improve patient outcomes and reduce the burden of liver disease.
Collapse
Affiliation(s)
- Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Michael Charlton
- Transplantation Institute, University of Chicago Medicine, Chicago, IL, USA
| | - Kenneth Cusi
- Division of Endocrinology, University of Florida, Gainesville, FL, USA
| | | | - Kris V Kowdley
- Liver Institute Northwest and Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| | - Mazen Noureddin
- Houston Research Institute and Houston Liver Institute, Houston, TX, USA
| | - Jay H Shubrook
- Department of Clinical Sciences and Community Health, Touro University California, Vallejo, CA, USA
| |
Collapse
|
39
|
Lin R, Zhou J, Sun Q, Xin X, Hu Y, Zheng M, Feng Q. Meta-analysis: Efficacy and safety of fibroblast growth factor 21 analogues for the treatment of non-alcoholic steatohepatitis and non-alcoholic steatohepatitis-related fibrosis. Aliment Pharmacol Ther 2024; 59:802-811. [PMID: 38297816 DOI: 10.1111/apt.17889] [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: 11/23/2023] [Revised: 12/18/2023] [Accepted: 01/13/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) analogues have emerged as promising therapeutic targets for non-alcoholic steatohepatitis (NASH). However, the effects and safety of these analogues on NASH and NASH-related fibrosis remain unexplored. AIMS To estimate the efficacy and safety of FGF21 analogues for treating NASH and NASH-related fibrosis. METHODS PubMed, Embase, and the Cochrane Library were searched for relevant studies up to 11 October 2023. Primary outcomes were defined as the fibrosis improvement ≥1 stage without worsening of NASH and NASH resolution without worsening fibrosis. Secondary outcomes included biomarkers of fibrosis, liver injury, and metabolism. Treatment-related adverse events were also analysed. RESULTS Nine studies, including 1054 patients with biopsy-proven NASH and stage F1-F4 fibrosis, were identified. Seven studies reported histological outcomes. The relative risk (RR) for obtaining fibrosis improvement ≥1 stage efficacy was 1.79 (95% CI 1.29-2.48, I2 = 37%, p < 0.001) with FGF21 analogues relative to placebo. Although no statistically significant difference was observed between FGF21 analogues in NASH resolution, sensitivity analyses and fragility index suggest that this result is unstable. The drugs improved hepatic fat fraction (HFF), along with other biomarkers of fibrosis, liver injury, and metabolism (MRE, LSM, Pro-C3, ELF, ALT, AST, TG, HDL-C, and LDL-C). Additionally, no significant difference in serious adverse event incidence rate was observed (RR = 1.26, 95% CI 0.82-1.94, I2 = 24%, p = 0.3). CONCLUSIONS FGF21 analogues appear as promising agents for the treatment of NASH and NASH-related fibrosis, and they generally seem to be safe and well tolerated.
Collapse
Affiliation(s)
- Rutao Lin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Clinical Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianghua Zhou
- Department of Cardiovascular Medicine, the Heart Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinmei Sun
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Xin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Minghua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Clinical Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Mandal M, Ghosh S, Roy S, Mandal S, Dasgupta A. Association of Diabetic Retinopathy with Midlife Hepatic Steatosis Diagnosed by Elastography and Hepatic Steatosis Index in Type 2 Diabetes in an Indian Population. Metab Syndr Relat Disord 2024; 22:214-221. [PMID: 38417047 DOI: 10.1089/met.2023.0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2024] Open
Abstract
Aims: People with type 2 diabetes mellitus are at increased risk of developing hepatic steatosis. We determined the prevalence of hepatic steatosis in middle-aged patients with and without diabetic retinopathy (DR) in an Indian population. We feel this information is critical, with trends of increasing chronic liver disease-related mortality at younger ages. Method: Institution-based analytical cross-sectional study with 114 middle-aged type 2 diabetes patients; 57 in each group with <15 years of duration of DM and without excessive drinking. Hepatic steatosis was determined by the hepatic steatosis index (HSI), hepatic ultrasonography (USG), and elastography. Result: The HSI in DR (37.9 ± 3.9) was more (P = 0.012) than in without diabetic retinopathy (NODR) (36.3 ± 3.3). There was no difference between two groups in liver span (P = 0.829) or in the prevalence of fatty liver (P = 0.562) as determined by conventional USG. Elastography value (kPa) was more (P = 0.001) in DR (6.51 ± 1.85) than in NODR (5.14 ± 1.60). On elastography, 50.9% in DR had a likelihood ratio (Metavir score for a stiffness value) for stage 2 Metavir score. In DR, 11.8% of those missed by USG had a likelihood ratio for ≥ stage 2 Metavir score on elastography. The presence of DR was independently correlated with kPa value (P < 0.001). Conclusion: A significantly higher prevalence of hepatic steatosis was observed in DR in this population. DR can be a useful biomarker for early hepatic screening in midlife, particularly with hepatic elastography, so that timely diagnosis of hepatic steatosis can be made.
Collapse
Affiliation(s)
- Mily Mandal
- Department of Ophthalmology, Calcutta National Medical College, Kolkata, India
| | - Sambuddha Ghosh
- Department of Ophthalmology, Calcutta National Medical College, Kolkata, India
| | - Satarupa Roy
- Department of Radiodiagnosis, Calcutta National Medical College, Kolkata, India
| | - Sayani Mandal
- Department of Radiodiagnosis, Calcutta National Medical College, Kolkata, India
| | - Anindya Dasgupta
- Department of Biochemistry, Jhargram Government Medical College, Jhargram, India
| |
Collapse
|
41
|
Shen X, Chen M, Zhang J, Lin Y, Gao X, Tu J, Chen K, Zhu A, Xu S. Unveiling the Impact of ApoF Deficiency on Liver and Lipid Metabolism: Insights from Transcriptome-Wide m6A Methylome Analysis in Mice. Genes (Basel) 2024; 15:347. [PMID: 38540406 PMCID: PMC10970566 DOI: 10.3390/genes15030347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024] Open
Abstract
Lipid metabolism participates in various physiological processes and has been shown to be connected to the development and progression of multiple diseases, especially metabolic hepatopathy. Apolipoproteins (Apos) act as vectors that combine with lipids, such as cholesterol and triglycerides (TGs). Despite being involved in lipid transportation and metabolism, the critical role of Apos in the maintenance of lipid metabolism has still not been fully revealed. This study sought to clarify variations related to m6A methylome in ApoF gene knockout mice with disordered lipid metabolism based on the bioinformatics method of transcriptome-wide m6A methylome epitranscriptomics. High-throughput methylated RNA immunoprecipitation sequencing (MeRIP-seq) was conducted in both wild-type (WT) and ApoF knockout (KO) mice. As a result, the liver histopathology presented vacuolization and steatosis, and the serum biochemical assays reported abnormal lipid content in KO mice. The m6A-modified mRNAs were conformed consensus sequenced in eukaryotes, and the distribution was enriched within the coding sequences and 3' non-coding regions. In KO mice, the functional annotation terms of the differentially expressed genes (DEGs) included cholesterol, steroid and lipid metabolism, and lipid storage. In the differentially m6A-methylated mRNAs, the functional annotation terms included cholesterol, TG, and long-chain fatty acid metabolic processes; lipid transport; and liver development. The overlapping DEGs and differential m6A-modified mRNAs were also enriched in terms of lipid metabolism disorder. In conclusion, transcriptome-wide MeRIP sequencing in ApoF KO mice demonstrated the role of this crucial apolipoprotein in liver health and lipid metabolism.
Collapse
Affiliation(s)
- Xuebin Shen
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping 353000, China; (X.S.); (Y.L.); (X.G.)
| | - Mengting Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (M.C.); (J.T.); (K.C.)
| | - Jian Zhang
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (M.C.); (J.T.); (K.C.)
| | - Yifan Lin
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping 353000, China; (X.S.); (Y.L.); (X.G.)
| | - Xinyue Gao
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping 353000, China; (X.S.); (Y.L.); (X.G.)
| | - Jionghong Tu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (M.C.); (J.T.); (K.C.)
| | - Kunqi Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (M.C.); (J.T.); (K.C.)
| | - An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (M.C.); (J.T.); (K.C.)
| | - Shanghua Xu
- Department of Cardiology, Affiliated Nanping First Hospital, Fujian Medical University, Nanping 353000, China; (X.S.); (Y.L.); (X.G.)
| |
Collapse
|
42
|
Nah EH, Choi YJ, Cho S, Park H, Kim S, Kwon E, Cho HI. Changes in nonalcoholic fatty liver disease and M2BPGi due to lifestyle intervention in primary healthcare. PLoS One 2024; 19:e0298151. [PMID: 38421976 PMCID: PMC10903829 DOI: 10.1371/journal.pone.0298151] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/06/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND A healthy lifestyle is the most important method for managing nonalcoholic fatty liver disease (NAFLD). Mac-2-binding protein glycosylated isomer (M2BPGi) has been suggested as a biomarker for NAFLD. This study aimed to determine the efficacy of personalized lifestyle interventions on NAFLD remission. METHODS This single-arm intervention study recruited participants with NAFLD who underwent health checkups at seven health-promotion centers in five South Korean cities. Fatty liver diagnosis was based on ultrasonography (US). The 109 individuals were recruited for personalized lifestyle interventions of hypocaloric diets and exercise. The participants attended the lifestyle intervention programs once per month for the first 3 months, and once every 3 months for the subsequent 6 months. In addition to sessions through center visits, phone-based intervention and self-monitoring at 4-, 5-, 7-, and 8-month were provided during the 9-month intervention period. And phone-based self-monitoring were also provided monthly during the 3-month follow-up period. The primary outcome was NAFLD remission at month 12 as measured on US and magnetic resonance elastography. The secondary outcomes were the changes in metabolic factors and M2BPGi. RESULTS The 108 individuals (62 males and 46 females; age 51.1±12.4 years, mean±standard deviation) were finally analyzed after the 12month intervention. Body mass index, waist circumference (WC), blood pressure, blood lipids (total cholesterol, triglycerides, and HDL-C), and fasting blood sugar levels were improved relative to baseline (all P<0.05). Fatty liver at or above the moderate grade according to US was decreased at month 12 relative to baseline (67.6% vs 50.9%) (P = 0.002). M2BPGi levels decreased during the 12-month study period (P<0.001). M2BPGi levels were moderately correlated with hepatic fat fraction by magnetic resonance imaging (r = 0.33, P = 0.05). WC (OR = 0.82, 95% CI = 0.67-1.00, P = 0.05) and HDL-C (OR = 1.17, 95% CI = 1.03-1.32, P = 0.014) were associated with remission of fatty liver in the multivariate analysis. CONCLUSION The personalized lifestyle intervention was effective in improving fatty liver and metabolic factors, but not hepatic stiffness, in NAFLD. TRIAL REGISTRATION ICTRP, cris.nih.go.kr (KCT0006380).
Collapse
Affiliation(s)
- Eun-Hee Nah
- Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Yong Jun Choi
- Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Seon Cho
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, South Korea
| | - Hyeran Park
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, South Korea
| | - Suyoung Kim
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, South Korea
| | - Eunjoo Kwon
- Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, South Korea
| | - Han-Ik Cho
- MEDIcheck LAB, Korea Association of Health Promotion, Seoul, South Korea
| |
Collapse
|
43
|
Wang M, Tang S, Li G, Huang Z, Mo S, Yang K, Chen J, Du B, Xu J, Ding Z, Dong F. Comparative study of ultrasound attenuation analysis and controlled attenuation parameter in the diagnosis and grading of liver steatosis in non-alcoholic fatty liver disease patients. BMC Gastroenterol 2024; 24:81. [PMID: 38395765 PMCID: PMC10885558 DOI: 10.1186/s12876-024-03160-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
PURPOSE To assess the diagnostic performance of Ultrasound Attenuation Analysis (USAT) in the diagnosis and grading of hepatic steatosis in patients with non-alcoholic fatty liver disease (NAFLD) using Controlled Attenuation Parameters (CAP) as a reference. MATERIALS AND METHODS From February 13, 2023, to September 26, 2023, participants underwent CAP and USAT examinations on the same day. We used manufacturer-recommended CAP thresholds to categorize the stages of hepatic steatosis: stage 1 (mild) - 240 dB/m, stage 2 (moderate) - 265 dB/m, stage 3 (severe) - 295 dB/m. Receiver Operating Characteristic curves were employed to evaluate the diagnostic accuracy of USAT and determine the thresholds for different levels of hepatic steatosis. RESULTS Using CAP as the reference, we observed that the average USAT value increased with the severity of hepatic steatosis, and the differences in USAT values among the different hepatic steatosis groups were statistically significant (p < 0.05). There was a strong positive correlation between USAT and CAP (r = 0.674, p < 0.0001). When using CAP as the reference, the optimal cut-off values for diagnosing and predicting different levels of hepatic steatosis with USAT were as follows: the cut-off value for excluding the presence of hepatic steatosis was 0.54 dB/cm/MHz (AUC 0.96); for mild hepatic steatosis, it was 0.59 dB/cm/MHz (AUC 0.86); for moderate hepatic steatosis, it was 0.73 dB/cm/MHz (AUC 0.81); and for severe hepatic steatosis, it was 0.87 dB/cm/MHz (AUC 0.87). CONCLUSION USAT exhibits strong diagnostic performance for hepatic steatosis and shows a high correlation with CAP values.
Collapse
Affiliation(s)
- Mengyun Wang
- The Second Clinical Medical College, Jinan University, Guangzhou, China
| | - Shuzhen Tang
- The Second Clinical Medical College, Jinan University, Guangzhou, China
| | - Guoqiu Li
- The Second Clinical Medical College, Jinan University, Guangzhou, China
| | - Zhibin Huang
- The Second Clinical Medical College, Jinan University, Guangzhou, China
| | - Sijie Mo
- The Second Clinical Medical College, Jinan University, Guangzhou, China
| | - Keen Yang
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Jing Chen
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Baishan Du
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Jinfeng Xu
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.
| | - Zhimin Ding
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.
| | - Fajin Dong
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.
| |
Collapse
|
44
|
Adeghate EA. GLP-1 receptor agonists in the treatment of diabetic non-alcoholic steatohepatitis patients. Expert Opin Pharmacother 2024; 25:223-232. [PMID: 38458647 DOI: 10.1080/14656566.2024.2328796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/06/2024] [Indexed: 03/10/2024]
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD) is the most common hepatic disease affecting almost 30% of the world population. Approximately 25% of people with NAFLD develop nonalcoholic steatohepatitis (NASH), the fulminant version of the disease. Diabetes mellitus is present in 22.5% of people with NAFLD and 44.60% of individuals with NASH. This review was undertaken to examine the current contribution of glucagon-like peptide 1 (GLP-1) receptor agonists to the pharmacotherapy of diabetic nonalcoholic steatohepatitis. AREAS COVERED The author analyzed the current status of GLP-1 receptor agonists for pharmacotherapy of diabetic NASH. Research data and literature reports were taken from the database and or websites of Diabetes UK, American Diabetes Association, ClinicalTrials.gov, PubMed, and Scopus. The keywords utilized included type 2 diabetes, GLP-1, NASH, NAFLD, and clinical trials. EXPERT OPINION Since diabetic NASH is associated with obesity, diabetes mellitus, oxidative stress and inflammation, drugs capable of mitigating all of these conditions simultaneously, are most ideal for the treatment of diabetic NASH. These drugs include (in order of relevance), GLP-1 receptor agonists, GLP-1 and GIP dual receptor agonists, sodium-glucose co-transporter-2 (SGLT2) inhibitors, and pioglitazone. The future, FDA-approved drug for diabetic NASH treatment will likely be GLP-1 agonist, which could be used as monotherapy or in combination with other drugs.
Collapse
Affiliation(s)
- Ernest A Adeghate
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
45
|
Sweeney JR, Arenas DJ, Fortuna D, Tondon R, Furth EE. Virtual biopsies: Proof of concept for a novel quantitative approach to liver biopsy adequacy and pathology education. Am J Clin Pathol 2024; 161:24-34. [PMID: 37598345 DOI: 10.1093/ajcp/aqad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023] Open
Abstract
OBJECTIVES To quantitatively measure liver biopsy adequacy requirements and the effect of a teaching intervention that uses a virtual biopsy platform. METHODS A library of virtual liver biopsies was created using digital whole-slide, trichrome-stained tissue sections from liver resection material and QuPath image analysis software. Blinded participants staged fibrosis on the virtual biopsies before and after a teaching intervention. RESULTS This platform both modeled adequacy requirements for cirrhosis diagnosis on biopsy material and measured the effect of a teaching intervention on participant performance. Using this platform, diagnostic accuracy for cirrhosis could be modeled according to the function y = λ(1 ‒ e‒x/γ). The platform demonstrated that the relationship between biopsy size and diagnostic accuracy was statistically significant and that biopsies smaller than 6 mm long and 0.8 mm wide were insufficient to diagnosis cirrhosis. The platform also measured improvement in fibrosis staging accuracy among participants following a teaching intervention. CONCLUSIONS These results provide proof of concept for a virtual biopsy method by which outstanding questions in anatomic pathology can be addressed quantitatively using open source software. Future work is needed to validate these findings in clinical practice.
Collapse
Affiliation(s)
- Jacob R Sweeney
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, US
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, US
| | - Daniel J Arenas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, US
| | - Danielle Fortuna
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, US
| | - Rashmi Tondon
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, US
| | - Emma E Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, US
| |
Collapse
|
46
|
Li L, Liao B, Cai H, Zhang Y, Deng K, Chen Y, Chen M, Zhou X, Tang M, Dong Z, Feng ST. Quantitative assessment of inflammation and evaluation of spatial heterogeneity for non-alcoholic fatty liver disease in mice based on iron-adjustive T1. Quant Imaging Med Surg 2024; 14:219-230. [PMID: 38223091 PMCID: PMC10784091 DOI: 10.21037/qims-23-782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/12/2023] [Indexed: 01/16/2024]
Abstract
Background A sensitive and non-invasive method is necessary to diagnose non-alcoholic fatty liver disease (NAFLD). We explored the iron-adjustive T1 (aT1) ability to quantify the degree of liver inflammation and evaluate the spatial heterogeneity. Methods Male C57BL/6J mice were randomly categorized as the NAFLD model (n=40), NAFLD-related liver cirrhosis model (n=20), and normal mice (n=10). T1 and T2* maps were acquired using a 3.0T scanner of magnetic resonance imaging (MRI) and aT1 maps through post-processing corrected iron's effect on T1 using T2*. Pathological changes in the left and right liver lobes were assessed using the Non-alcoholic Steatohepatitis-Clinical Research Network scoring system, though hepatic ballooning lesion were rare in models. Spearman's and partial correlation analyses were used to evaluate correlations, and the receiver operating characteristic curve was used to analyze the diagnostic performance. Results aT1 was highly correlated with NAFLD activity score (NAS) (r=0.747, P<0.001) but not with the fibrosis stage when adjusted by NAS (r=-0.135, P=0.147). The area under the curve (AUC) of the aT1 value distinguishing groups with 0< NAS <4 and NAS ≥4 was 0.802. On analyzing the histogram features of aT1, the entropy, interquartile range, range, and variance were significantly different between the groups with 0< NAS <4 and NAS ≥4 (P<0.05). The entropy was the risk factor of NAS ≥4. Conclusions aT1 could help evaluate the inflammatory activity in NAFLD mice unaffected by mild fibrosis, and the higher the degree of inflammation, the higher the heterogeneity of the aT1 map.
Collapse
Affiliation(s)
- Lujie Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bing Liao
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huasong Cai
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yinhong Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kan Deng
- Philips Healthcare, Guangzhou, China
| | - Yuying Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meicheng Chen
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqi Zhou
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mimi Tang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi Dong
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
47
|
Yang S, Zhou L, Zhao T, Zhu H, Luo T, Jiang K, Shi X, Chen C, Zhang H, Zhao S, Zou X, Zhuge Y, Wang F, Wang L, Jiang M, Xu B. Protective and Adverse Roles of DDX3X in Different Cell Types in Nonalcoholic Steatohepatitis Progression. RESEARCH (WASHINGTON, D.C.) 2023; 6:0275. [PMID: 38090607 PMCID: PMC10712874 DOI: 10.34133/research.0275] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/09/2023] [Indexed: 04/19/2025]
Abstract
Persistent hepatic cellular metabolic stress and liver inflammatory stimuli are key signatures of nonalcoholic steatohepatitis (NASH). DDX3X is a vital molecule involved in cell fate decisions in both pro-survival stress granule (SG) and pro-death NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome assembly in response to stress signals. However, the role of DDX3X in NASH remains unclear. We characterized the cell type-specific roles of DDX3X in NASH. Human liver tissues from NASH patients and normal control subjects were collected to assess DDX3X expression and distribution. Nutritional steatohepatitis models were constructed by feeding macrophage-specific DDX3X knockout (DDX3XΔMφ), hepatocyte-specific DDX3X knockout (DDX3XΔhep), and wild-type control (DDX3Xfl/fl) mice a high-fat and high-cholesterol (HFHC) diet, a methionine- and choline-deficient (MCD) diet, and a high-fat/high-iron/high-fructose/high-cholesterol, low-methionine, and choline-deficient (HFHIHFHC-MCD) diet. The study demonstrated that DDX3X was predominantly expressed in macrophages and hepatocytes in control liver tissues, and its expression was down-regulated in patients or mice with NASH. Compared to DDX3Xfl/fl littermates, DDX3XΔMφ mice showed improved liver histology in nutritional steatohepatitis models. Loss of macrophage DDX3X inhibited NLRP3 inflammasome-mediated pyroptosis, causing anti-inflammatory M2 polarization and alleviating hepatocyte steatohepatitic changes. DDX3XΔhep mice developed marked steatohepatitis in multiple nutritional steatohepatitis models compared to DDX3Xfl/fl littermates. DDX3X-deleted hepatocytes showed impaired SG assembly, leading to increased sensitivity and intolerance to metabolic stimulation and resultant steatohepatitis. In conclusion, DDX3X plays opposite roles in different cell types during the progression of NASH. A better understanding of the cell-specific differences in the crosstalk between SG formation and NLRP3 activation is crucial for developing prospective targeted DDX3X inhibitors for the treatment of NASH.
Collapse
Affiliation(s)
- Suzhen Yang
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Lin Zhou
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Tianming Zhao
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
- Department of Gastroenterology,
Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Jiangsu, Nanjing, 210008, China
| | - Hanlong Zhu
- Department of Gastroenterology and Hepatology,
the Affiliated Jinling Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Tingting Luo
- School of Medicine,
Northwest University, Shaanxi, Xi’an, 710069, China
| | - Kang Jiang
- Department of Gastroenterology and Hepatology,
the Affiliated Jinling Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Xiaoxiao Shi
- Department of Gastroenterology and Hepatology,
the Affiliated Jinling Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Chunyan Chen
- Department of Gastroenterology and Hepatology,
the Affiliated Jinling Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Han Zhang
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Si Zhao
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Xiaoping Zou
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Yuzheng Zhuge
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Fangyu Wang
- Department of Gastroenterology and Hepatology,
the Affiliated Jinling Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Lei Wang
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Mingzuo Jiang
- Department of Gastroenterology and Hepatology,
the Affiliated Jinling Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| | - Bing Xu
- Department of Gastroenterology,
the Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu, Nanjing, 210002, China
| |
Collapse
|
48
|
Lin H, Qiu S, Yang Y, Yang C, Shen Z, Chen Y, Zhang Z, Feng Y, Yan F. Three-dimensional magnetic resonance elastography combining proton-density fat fraction precisely identifies metabolic dysfunction-associated steatohepatitis with significant fibrosis. Magn Reson Imaging 2023; 104:1-8. [PMID: 37553044 DOI: 10.1016/j.mri.2023.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/10/2023]
Abstract
PURPOSE Patients with metabolic dysfunction-associated steatohepatitis (MASH) and significant fibrosis (fibrosis stage≥2), known as Fibro-MASH, are at increased risk of liver-related outcomes and lower rates of spontaneous disease regression. The aim was to investigate three-dimensional MR elastography (3D-MRE) combining proton-density fat fraction (PDFF) as a means of identifying Fibro-MASH. METHODS Forty-eight New Zealand rabbits were fed a high-fat/cholesterol or standard diet to obtain different disease activity and fibrosis stages. Shear stiffness (SS) and Damping Ratio (DR) were derived from 3D-MRE, whereas PDFF was from a volumetric 3D imaging sequence. Steatosis grade, metabolic dysfunction-associated steatotic liver disease activity score (MAS), and fibrosis stage were diagnosed histologically. Serum markers of fibrosis and inflammation were also measured. Correlation and comparison analysis, Receiver operating characteristic curves (ROC), Delong test, logistic regression analysis, and Net reclassification improvement (NRI) were performed. RESULTS PDFF correlated with steatosis grade (rho = 0.853). SS increased with developed liver fibrosis (rho = 0.837). DR correlated with MAS grade (rho = 0.678). The areas under the ROC (AUROCs) of SS for fibrosis grading were 0.961 and 0.953 for ≥F2, and ≥ F3, respectively. All the biochemical parameters were considered but excluded from the logistic regression analysis to identify Fibro-MASH. FF, SS, and DR were finally included in the further analysis. The three-parameter model combining PDFF, SS, and DR showed significant improvement in NRI over the model combining SS and PDFF (AUROC 0.973 vs. 0.906, P = 0.081; NRI 0.28, P < 0.05). CONCLUSION 3D-MRE combining PDFF may characterize the state of fat content, disease activity and fibrosis, thus precisely identify Fibro-MASH.
Collapse
Affiliation(s)
- Huimin Lin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Suhao Qiu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Yanzhao Yang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chunxue Yang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhehan Shen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yu Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhihan Zhang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuan Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
49
|
Dichtel LE, Corey KE, Haines MS, Chicote ML, Lee H, Kimball A, Colling C, Simon TG, Long MT, Husseini J, Bredella MA, Miller KK. Growth Hormone Administration Improves Nonalcoholic Fatty Liver Disease in Overweight/Obesity: A Randomized Trial. J Clin Endocrinol Metab 2023; 108:e1542-e1550. [PMID: 37379033 PMCID: PMC10655511 DOI: 10.1210/clinem/dgad375] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023]
Abstract
CONTEXT Overweight and obesity are associated with relative growth hormone (GH) deficiency, which has been implicated in the development of nonalcoholic fatty liver disease (NAFLD). NAFLD is a progressive disease without effective treatments. OBJECTIVE We hypothesized that GH administration would reduce hepatic steatosis in individuals with overweight/obesity and NAFLD. METHODS In this 6-month randomized, double-blind, placebo-controlled trial of low-dose GH administration, 53 adults aged 18 to 65 years with BMI ≥25 kg/m2 and NAFLD without diabetes were randomized to daily subcutaneous GH or placebo, targeting insulin-like growth factor 1 (IGF-1) to the upper normal quartile. The primary endpoint was intrahepatic lipid content (IHL) by proton magnetic resonance spectroscopy (1H-MRS) assessed before treatment and at 6 months. RESULTS Subjects were randomly assigned to a treatment group (27 GH; 26 placebo), with 41 completers (20 GH and 21 placebo) at 6 months. Reduction in absolute % IHL by 1H-MRS was significantly greater in the GH vs placebo group (mean ± SD: -5.2 ± 10.5% vs 3.8 ± 6.9%; P = .009), resulting in a net mean treatment effect of -8.9% (95% CI, -14.5 to -3.3%). All side effects were similar between groups, except for non-clinically significant lower extremity edema, which was more frequent in the GH vs placebo group (21% vs 0%, P = .02). There were no study discontinuations due to worsening of glycemic status, and there were no significant differences in change in glycemic measures or insulin resistance between the GH and placebo groups. CONCLUSION GH administration reduces hepatic steatosis in adults with overweight/obesity and NAFLD without worsening glycemic measures. The GH/IGF-1 axis may lead to future therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Laura E Dichtel
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Kathleen E Corey
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Melanie S Haines
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Mark L Chicote
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Allison Kimball
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Caitlin Colling
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Tracey G Simon
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Michelle T Long
- Section of Gastroenterology, Boston Medical Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jad Husseini
- Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Miriam A Bredella
- Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Karen K Miller
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
50
|
Xiang S, Li Y, Li Y, Pan W, Wang X, Lu Y, Liu S. Higher anthocyanin intake is associated with a lower risk of non-alcoholic fatty liver disease in the United States adult population. Front Nutr 2023; 10:1265507. [PMID: 38024364 PMCID: PMC10657849 DOI: 10.3389/fnut.2023.1265507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background Flavonoids are a class of plant chemicals known to have health-promoting properties, including six subclasses. Anthocyanin is one of the subclasses that have anti-inflammatory and antioxidant activities. However, the relationship between flavonoid subclass intake and the risk of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis has not been verified in representative samples of the United States. Methods This is a cross-sectional study based on the data from the National Health and Nutrition Examination Survey (NHANES) and the Food and Nutrient Database for Dietary Studies (FNDDS) in 2017-2018. The intake of flavonoid subclasses of the participants was obtained from two 24 h dietary recalls. The NAFLD and liver fibrosis were defined based on the international consensus criteria. The relationship between flavonoid subclass intake and NAFLD and liver fibrosis was evaluated using a multivariate logistic regression model corrected for multiple confounding factors. Subgroup analysis, trend tests, interaction tests and restricted cubic spline were carried out to further explore this relationship. In addition, we also explored the relationship between anthocyanin and liver serum biomarkers, dietary total energy intake and healthy eating index (HEI)-2015 scores. Results A total of 2,288 participants were included in the analysis. The intake of anthocyanin was significantly negatively associated with the risk of NAFLD, but not other flavonoid subclasses. A higher anthocyanin intake was significantly associated with a lower risk of NAFLD (quartile 4, OR 0.470, 95% CI 0.275-0.803). The results of subgroup analysis showed that the protective effect of dietary anthocyanin intake on NAFLD was more pronounced in participants of non-Hispanic whites, with hypertension and without diabetes (P for interaction <0.05). Alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), dietary total energy intake was significantly negatively correlated with dietary anthocyanin intake. We did not find any protective effect of flavonoid subclass intake on liver fibrosis. Conclusion Anthocyanin, but not other flavonoid subclasses, can significantly reduce the risk of NAFLD. The protective effect was more pronounced in non-Hispanic whites, participants without diabetes and those with hypertension. Our study provides new evidence that anthocyanin intake has a reverse significant association with the risk for NAFLD.
Collapse
Affiliation(s)
- Shuai Xiang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujing Li
- College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Pathology, The First Hospital of China Medical University, Shenyang, China
| | - Ying Li
- Department of Blood Transfusion, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjun Pan
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoqian Wang
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yun Lu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shanglong Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|