1
|
Yang CY, Chen JH, Chen CY, Kao CY, Huang SF, Chang WY, Tu HP, Huang JF, Yu ML, Tai CM. Serial changes in metabolic dysfunction-associated steatotic liver disease after sleeve gastrectomy and their associations with abdominal adiposity: a prospective cohort study. Surg Obes Relat Dis 2025; 21:537-546. [PMID: 39706718 DOI: 10.1016/j.soard.2024.11.007] [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: 07/09/2024] [Revised: 10/10/2024] [Accepted: 11/13/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Little is known about the associations between changes in hepatic steatosis and changes in abdominal adiposity after metabolic bariatric surgery. OBJECTIVES To evaluate the serial changes in hepatic steatosis and abdominal adiposity following sleeve gastrectomy (SG). SETTING University hospital, Taiwan. METHODS In this prospective study, patients who underwent SG and intraoperative liver biopsy were enrolled. Magnetic resonance imaging (MRI) was performed to assess the liver fat fraction (LFF), visceral adipose tissue (VAT) area, and subcutaneous adipose tissue (SAT) area. Liver fibrosis was assessed preoperatively via biopsy and the fibrosis-4 index (FIB-4) and postoperatively with the FIB-4. RESULTS Seventy-six metabolic dysfunction-associated steatotic liver disease (MASLD) patients, including 67 pure MASLD patients and 9 MASLD patients with combined etiologies, were enrolled. LFF and visceral-to-subcutaneous fat ratio were associated with metabolic dysfunction-associated steatohepatitis, and VAT area was associated with significant fibrosis (≥F2). Twelve months after SG, all MRI measurements significantly improved. The median LFF of pure MASLD patients decreased from 17.4% at baseline to 4.2% and 3.7% at the 6th and 12th postoperative months, respectively. Complete resolution of steatosis was achieved in 97.5% of patients at the 12th postoperative months. Using %VAT and %SAT reductions at the sixth postoperative month as references, LFF decreased more rapidly, with fold ratios of 1.3 and 1.8, respectively. CONCLUSIONS SG resulted in a significant decrease in hepatic steatosis and abdominal adiposity in patients with severe obesity, but hepatic steatosis improved faster than abdominal adiposity. Hepatic steatosis resolved in almost all patients 12 months after SG.
Collapse
Affiliation(s)
- Chung-Yi Yang
- Department of Medical imaging, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan
| | - Jian-Han Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; Division of General Surgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Chung-Yen Chen
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; Division of General Surgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Cheng-Yi Kao
- Department of Medical imaging, E-DA Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Shiu-Feng Huang
- Investigator and Attending Physician, Institute of Molecular and Genomic Medicine, National Health Research Institutes
| | - Wen-Yu Chang
- Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; Department of Dermatology, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Pin Tu
- Department of Public Health and Environmental Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center of Hepatitis Research, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Center of Hepatitis Research, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chi-Ming Tai
- Bariatric and Metabolism International Surgery Center, E-Da Hospital, Kaohsiung, Taiwan; School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.
| |
Collapse
|
2
|
Huang Q, Qadri SF, Bian H, Yi X, Lin C, Yang X, Zhu X, Lin H, Yan H, Chang X, Sun X, Ma S, Wu Q, Zeng H, Hu X, Zheng Y, Yki-Järvinen H, Gao X, Tang H, Xia M. A metabolome-derived score predicts metabolic dysfunction-associated steatohepatitis and mortality from liver disease. J Hepatol 2025; 82:781-793. [PMID: 39423864 DOI: 10.1016/j.jhep.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 09/11/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) is associated with a >10-fold increase in liver-related mortality. However, biomarkers predicting both MASH and mortality in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) are missing. We developed a metabolome-derived prediction score for MASH and examined whether it predicts mortality in Chinese and European cohorts. METHODS The MASH prediction score was developed using a multi-step machine learning strategy, based on 44 clinical parameters and 250 serum metabolites measured by proton nuclear magnetic resonance in 311 Chinese adults undergoing a liver biopsy. External validation was conducted in a Finnish liver biopsy cohort (n = 305). We investigated associations of the score with all-cause and cause-specific mortality in the population-based Shanghai Changfeng study (n = 5,893) and the UK biobank (n = 111,673). RESULTS A total of 24 clinical parameters and 194 serum metabolites were significantly associated with MASH in the Chinese liver biopsy cohort. The final MASH score included BMI, aspartate aminotransferase, tyrosine, and the phospholipid-to-total lipid ratio in VLDL. The score identified patients with MASH with AUROCs of 0.87 (95% CI 0.83-0.91) and 0.81 (95% CI 0.75-0.88) in the Chinese and Finnish cohorts, with high negative predictive values. Participants with a high or intermediate risk of MASH based on the score had a markedly higher risk of MASLD-related mortality than those with a low risk in Chinese (hazard ratio 23.19; 95% CI 4.80-111.97) and European (hazard ratio 20.15; 95% CI 10.95-37.11) individuals after 7.2 and 12.6 years of follow-up, respectively. The MASH prediction score was superior to the Fibrosis-4 index and the NAFLD fibrosis score in predicting MASLD-related mortality. CONCLUSION The metabolome-derived MASH prediction score accurately predicts risk of MASH and MASLD-related mortality in both Chinese and European individuals. IMPACT AND IMPLICATIONS Metabolic dysfunction-associated steatohepatitis (MASH) is associated with more than a 10-fold increase in liver-related death. However, biomarkers predicting not only MASH, but also death due to liver disease, are missing. We established a MASH prediction score based on 44 clinical parameters and 250 serum metabolites using a machine learning strategy. This metabolome-derived MASH prediction score could accurately identify patients with MASH among both Chinese and Finnish individuals, and it was superior to the Fibrosis-4 index and the NAFLD fibrosis score in predicting MASLD-related death in the general population. Thus, the new MASH prediction score is a useful tool for identifying individuals with a markedly increased risk of serious liver-related outcomes among at-risk and general populations.
Collapse
Affiliation(s)
- Qingxia Huang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Sami F Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiaoxuan Yi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Chenhao Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Xinyu Yang
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiaopeng Zhu
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xinxia Chang
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiaoyang Sun
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Shuai Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Qi Wu
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Hailuan Zeng
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | - Xiqi Hu
- Department of Pathology, Medical College, Fudan University, Shanghai, China
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Zhongshan Hospital, Fudan University, Shanghai 200438, China.
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Rahim M, Bednarski TK, Hasenour CM, Banerjee DR, Trenary I, Young JD. Simultaneous in vivo multi-organ fluxomics reveals divergent metabolic adaptations in liver, heart, and skeletal muscle during obesity. Cell Rep 2025; 44:115591. [PMID: 40244853 DOI: 10.1016/j.celrep.2025.115591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 02/23/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
We present an isotope-based metabolic flux analysis (MFA) approach to simultaneously quantify metabolic fluxes in the liver, heart, and skeletal muscle of individual mice. The platform was scaled to examine metabolic flux adaptations in age-matched cohorts of mice exhibiting varying levels of chronic obesity. We found that severe obesity increases hepatic gluconeogenesis and citric acid cycle flux, accompanied by elevated glucose oxidation in the heart that compensates for impaired fatty acid oxidation. In contrast, skeletal muscle fluxes exhibit an overall reduction in substrate oxidation. These findings demonstrate the dichotomy in fuel utilization between cardiac and skeletal muscle during worsening metabolic disease and demonstrate the divergent effects of obesity on metabolic fluxes in different organs. This multi-tissue MFA technology can be extended to address important questions about in vivo regulation of metabolism and its dysregulation in disease, which cannot be fully answered through studies of single organs or isolated cells/tissues.
Collapse
Affiliation(s)
- Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
4
|
Onoiu AI, Domínguez DP, Joven J. Digital Pathology Tailored for Assessment of Liver Biopsies. Biomedicines 2025; 13:846. [PMID: 40299404 PMCID: PMC12024806 DOI: 10.3390/biomedicines13040846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Improved image quality, better scanners, innovative software technologies, enhanced computational power, superior network connectivity, and the ease of virtual image reproduction and distribution are driving the potential use of digital pathology for diagnosis and education. Although relatively common in clinical oncology, its application in liver pathology is under development. Digital pathology and improving subjective histologic scoring systems could be essential in managing obesity-associated steatotic liver disease. The increasing use of digital pathology in analyzing liver specimens is particularly intriguing as it may offer a more detailed view of liver biology and eliminate the incomplete measurement of treatment responses in clinical trials. The objective and automated quantification of histological results may help establish standardized diagnosis, treatment, and assessment protocols, providing a foundation for personalized patient care. Our experience with artificial intelligence (AI)-based software enhances reproducibility and accuracy, enabling continuous scoring and detecting subtle changes that indicate disease progression or regression. Ongoing validation highlights the need for collaboration between pathologists and AI developers. Concurrently, automated image analysis can address issues related to the historical failure of clinical trials stemming from challenges in histologic assessment. We discuss how these novel tools can be incorporated into liver research and complement post-diagnosis scenarios where quantification is necessary, thus clarifying the evolving role of digital pathology in the field.
Collapse
Affiliation(s)
- Alina-Iuliana Onoiu
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, 43204 Reus, Spain;
- Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - David Parada Domínguez
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, 43204 Reus, Spain;
- Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, 43201 Reus, Spain
- Department of Pathology, Hospital Universitari Sant Joan, 43204 Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan, Universitat Rovira i Virgili, 43204 Reus, Spain;
- Department of Medicine and Surgery, Faculty of Medicine, Universitat Rovira i Virgili, 43201 Reus, Spain
- The Campus of International Excellence Southern Catalonia, 43003 Tarragona, Spain
| |
Collapse
|
5
|
Cathcart J, Barrett R, Bowness JS, Mukhopadhya A, Lynch R, Dillon JF. Accuracy of Non-Invasive Imaging Techniques for the Diagnosis of MASH in Patients With MASLD: A Systematic Review. Liver Int 2025; 45:e16127. [PMID: 39400428 PMCID: PMC11891385 DOI: 10.1111/liv.16127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/14/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health problem. The secondary stage in MASLD is steatohepatitis (MASH), the co-existence of steatosis and inflammation, a leading cause of progression to fibrosis and mortality. MASH resolution alone improves survival. Currently, MASH diagnosis is via liver biopsy. This study sought to evaluate the accuracy of imaging-based tests for MASH diagnosis, which offer a non-invasive method of diagnosis. METHODS Eight academic literature databases were searched and references of previous systematic reviews and included papers were checked for additional papers. Liver biopsy was used for reference standard. RESULTS We report on 69 imaging-based studies. There were 31 studies on MRI, 27 on ultrasound, five on CT, 13 on transient elastography, eight on controlled attenuation parameter (CAP) and two on scintigraphy. The pathological definition of MASH was inconsistent, making it difficult to compare studies. 55/69 studies (79.71%) were deemed high-risk of bias as they had no preset thresholds and no validation. The two largest groups of imaging papers were on MRI and ultrasound. AUROCs were up to 0.93 for MRE, 0.90 for MRI, 1.0 for magnetic resonance spectroscopy (MRS) and 0.94 for ultrasound-based studies. CONCLUSIONS Our study found that the most promising imaging tools are MRI techniques or ultrasound-based scores and confirmed there is potential to utilise these for MASH diagnosis. However, many publications are single studies without independent prospective validation. Without this, there is no clear imaging tool or score currently available that is reliably tested to diagnose MASH.
Collapse
Affiliation(s)
- Jennifer Cathcart
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
- Gastroenterology DepartmentAberdeen Royal InfirmaryAberdeenUK
| | - Rachael Barrett
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
| | - James S. Bowness
- University College London Hospitals NHS Foundation TrustLondonUK
- Department of Targeting InterventionUniversity College LondonLondonUK
| | | | - Ruairi Lynch
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
| | - John F. Dillon
- Division of Molecular and Clinical MedicineUniversity of DundeeDundeeUK
| |
Collapse
|
6
|
Fichez J, Mouillot T, Vonghia L, Costentin C, Moreau C, Roux M, Delamarre A, Francque S, Zheng MH, Boursier J. Non-invasive tests for fibrotic MASH for reducing screen failure in therapeutic trials. JHEP Rep 2025; 7:101351. [PMID: 40212791 PMCID: PMC11985113 DOI: 10.1016/j.jhepr.2025.101351] [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: 09/17/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND & AIMS Therapeutic trials in metabolic dysfunction-associated steatohepatitis (MASH) are hampered by a high 70-80% screen failure rate mostly because of the absence of fibrotic MASH on baseline liver biopsies, underscoring the need for better selection of candidates. We compared the performance of eight non-invasive tests, designed or not for the diagnosis of fibrotic MASH. METHODS A total of 1,005 patients with histologically proven MASLD were included in five tertiary care centers. Three non-invasive tests developed for fibrotic MASH were evaluated: the simple blood test Fibrotic NASH Index (FNI), the specialized blood test MACK-3, and the elastography-based test FAST. Five non-invasive tests recommended for advanced fibrosis were evaluated as well: the simple blood test FIB-4, the specialized blood tests FibroTest and Enhanced Liver Fibrosis test (ELF™), and the elastography-based tests FibroScan and Agile3+. Fibrotic MASH was defined as MASH with MASLD activity score ≥4 and fibrosis score F ≥2. RESULTS Among simple blood tests (n = 1,005), FNI had a significantly higher area under the receiver operating characteristic (AUROC) for fibrotic MASH than FIB-4 (0.709 [0.677-0.741] vs. 0.662 [0.628-0.695], p = 0.019). Among elastography-based tests (n = 817), FAST had a significantly higher AUROC (0.774 [0.743-0.806]) than FibroScan (0.728 [0.694-0.763], p = 0.013) and Agile3+ (0.708 [0.672-0.744], p = 0.004). Among specialized blood tests (n = 545), MACK-3 had a significantly higher AUROC (0.772 [0.734-0.811]) than FibroTest (0.615 [0.568-0.663], p <0.001) and ELF (0.700 [0.656-0.744], p = 0.028). Sequential combination (FAST then Agile3+; MACK-3 then ELF) identified a subset including one-third of patients in whom the false-positive rate was only 30%. CONCLUSIONS Sequential combinations using first-line tests designed for fibrotic MASH improves the identification of candidates for MASH therapeutic trials. IMPACT AND IMPLICATIONS Drug development in metabolic dysfunction-associated steatohepatitis (MASH) is hampered by a high screen failure rate, one of the main reasons being the absence of MASH and significant fibrosis (fibrotic MASH) on the baseline liver biopsy, a key inclusion criterion in MASH therapeutic trials. Non-invasive tests represent an attractive opportunity to better select candidates for these trials, but most of them have been developed for advanced fibrosis and the new tests designed for the diagnosis of fibrotic MASH remain poorly validated. In this work, we demonstrate that the tests specifically designed for fibrotic MASH are more accurate for this diagnostic target than the tests currently recommended and initially developed for advanced fibrosis. We propose sequential combinations that will facilitate the identification of patients with fibrotic MASH in need of treatment, and their inclusion in MASH therapeutic trials.
Collapse
Affiliation(s)
- Jeanne Fichez
- Hepato-Gastroenterology and Digestive Oncology Department, Angers University Hospital, Angers, France
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| | - Thomas Mouillot
- Hepato-Gastroenterology and Digestive Oncology Department, Dijon University Hospital, Dijon, France
| | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Charlotte Costentin
- Grenoble Alpes University/Hepato-Gastroenterology and Digestive Oncology Department, Grenoble Alpes University Hospital, Grenoble, France
- Grenoble Institute for Advanced Biosciences, Research Center UGA/Inserm U 1209/CNRS 5309, Grenoble Alpes University, Grenoble, France
| | - Clémence Moreau
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
- Department of Methodology and Biostatistics, Angers University Hospital, Angers, France
| | - Marine Roux
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| | - Adèle Delamarre
- Hepatology Unit, Haut Leveque Hospital, Bordeaux University Hospital, Bordeaux, France
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jérôme Boursier
- Hepato-Gastroenterology and Digestive Oncology Department, Angers University Hospital, Angers, France
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| |
Collapse
|
7
|
Park JH, Kim SM, Lee DH. Comparison of Non-invasive Methods for Diagnosis of Non-alcoholic Fatty Liver Disease Before Bariatric Surgery and Postoperative Follow-up in Obese Patients. JOURNAL OF METABOLIC AND BARIATRIC SURGERY 2025; 14:53-64. [PMID: 40351816 PMCID: PMC12059311 DOI: 10.17476/jmbs.2025.14.1.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Purpose This study aims to identify the most accurate and useful non-invasive method to replace liver biopsy for the diagnosis of non-alcoholic fatty liver disease (NAFLD) before bariatric surgery and postoperative follow-up in morbidly obese patients. Materials and Methods This single-center study is a retrospective analysis of prospectively collected data from 68 morbidly obese patients who underwent laparoscopic sleeve gastrectomy with intraoperative liver biopsy. Preoperative non-invasive diagnostic methods, including fatty liver index, NAFLD fibrosis score, enhanced liver fibrosis score, FibroScan, magnetic resonance imaging-proton density fat fraction (MRI-PDFF), magnetic resonance spectroscopy (MRS)-PDFF, and magnetic resonance elastography (MRE) were compared against liver biopsy results. Diagnostic performance was assessed using Spearman's correlation and receiver operating characteristic (ROC) curve analysis. Results Liver biopsy confirmed the presence of steatosis in 92.7% of patients, Nonalcoholic Steatohepatitis (NASH) in 64.7%, and liver fibrosis (≥F1) in 72.0%. MRI-PDFF and MRS-PDFF demonstrated the highest diagnostic accuracy for NASH, with the strongest correlation with histological findings. For liver fibrosis, MRE showed the strongest correlation with histological fibrosis stage, while FibroScan-Liver Stiffness Measurement (LSM) demonstrated better diagnostic performance in ROC analysis. However, the overall diagnostic quality of non-invasive methods for fibrosis assessment remained modest, with no method achieving a quality value above 0.6. Conclusion MRI-PDFF and MRS-PDFF were the most accurate noninvasive methods for diagnosing NASH in morbidly obese patients. For liver fibrosis, FibroScan-LSM may be more suitable for detection, while MRE may better reflect fibrosis severity. Further studies are needed to assess the cost-effectiveness and clinical applicability of these methods.
Collapse
Affiliation(s)
- Ji-Hyeon Park
- Department of Surgery, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Seong Min Kim
- Department of Surgery, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
8
|
Li XQ, Cheng GW, Akiyama I, Huang XJ, Liang J, Xue LY, Cheng Y, Kudo M, Ding H. Attenuation imaging: Diagnostic differences in hepatic steatosis for chronic hepatitis B vs metabolic dysfunction-associated steatotic liver disease patients. World J Gastroenterol 2025; 31:102795. [PMID: 40124278 PMCID: PMC11924003 DOI: 10.3748/wjg.v31.i11.102795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/25/2025] [Accepted: 02/20/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND Hepatic steatosis, characterized by fat accumulation in hepatocytes, can result from metabolic dysfunction-associated steatotic liver disease (MASLD), infections, alcoholism, chemotherapy, and toxins. MASLD is diagnosed via imaging or biopsy with metabolic criteria and may progress to metabolic dysfunction-associated steatohepatitis, potentially leading to fibrosis, cirrhosis, or cancer. The coexistence of hepatic steatosis with chronic hepatitis B (CHB) is mainly related to metabolic factors and increases mortality and cancer risks. As a noninvasive method, attenuation imaging (ATI) shows promise in quantifying liver fat, demonstrating strong correlation with liver biopsy. AIM To investigate the disparity of ATI for assessing biopsy-based hepatic steatosis in CHB patients and MASLD patients. METHODS The study enrolled 249 patients who underwent both ATI and liver biopsy, including 78 with CHB and 171 with MASLD. Hepatic steatosis was classified into grades S0 to S3 according to the proportion of fat cells present. Liver fibrosis was staged from 0 to 4 according to the meta-analysis of histological data in viral hepatitis scoring system. The diagnostic performance of attenuation coefficient (AC) values across different groups was compared for each grade of steatosis. Factors associated with the AC values were determined through linear regression analysis. A multivariate logistic regression model was established to predict ≥ S2 within the MASLD group. RESULTS In both the CHB and the MASLD groups, AC values increased significantly with higher steatosis grade (P < 0.001). In the CHB group, the areas under the curve (AUCs) of AC for predicting steatosis grades ≥ S1, ≥ S2 and S3 were 0.918, 0.960 and 0.987, respectively. In contrast, the MASLD group showed AUCs of 0.836, 0.774, and 0.688 for the same steatosis grades. The diagnostic performance of AC for detecting ≥ S2 and S3 indicated significant differences between the two groups (both P < 0.001). Multivariate linear regression analysis identified body mass index, triglycerides, and steatosis grade as significant factors for AC. When the steatosis grade is ≥ S2, it can progress to more serious liver conditions. A clinical model integrating blood biochemical parameters and AC was developed in the MASLD group to enhance the prediction of ≥ S2, achieving an AUC of 0.848. CONCLUSION The AC could effectively discriminate the degree of steatosis in both the CHB and MASLD groups. In the MASLD group, when combined with blood biochemical parameters, AC exhibited better predictive ability for moderate to severe steatosis.
Collapse
Affiliation(s)
- Xue-Qi Li
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Guang-Wen Cheng
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Iwaki Akiyama
- Medical Ultrasound Research Center, Doshisha University, Kyoto 600-8586, Kyōto, Japan
| | - Xian-Jue Huang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Liang
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Li-Yun Xue
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yi Cheng
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka 577-8502, Japan
| | - Hong Ding
- Department of Ultrasound, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
9
|
Boursier J, Hervé H, Roux M, Abdelmalek MF, Francque SM, Broqua P, Junien JL, Abitbol JL, Huot-Marchand P, Dzen L, Cooreman MP, Patel S. Biomarkers of Histological Response in Lanifibranor-treated Patients With Metabolic Dysfunction-associated Steatohepatitis. Clin Gastroenterol Hepatol 2025:S1542-3565(25)00204-6. [PMID: 40107637 DOI: 10.1016/j.cgh.2024.12.039] [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: 08/03/2024] [Revised: 12/01/2024] [Accepted: 12/23/2024] [Indexed: 03/22/2025]
Abstract
BACKGROUND & AIMS Lanifibranor, a pan-peroxisome proliferator-activated receptor agonist, has demonstrated therapeutic efficacy on metabolic dysfunction-associated steatohepatitis (MASH) resolution and fibrosis improvement in the Phase IIb NATIVE study. The histologic endpoints of MASH resolution and fibrosis improvement (E1), MASH resolution without worsening of fibrosis (E2), and fibrosis improvement without worsening of MASH (E3) were investigated with the aim of identifying biological signatures of E1, E2, and E3 responders based on serum biomarkers in patients treated with lanifibranor. METHODS NATIVE evaluated lanifibranor 800 and 1200 mg daily vs placebo in patients with non-cirrhotic MASH treated over 24 weeks. Liver biopsy was obtained at baseline and the end of treatment. Patients receiving lanifibranor were pooled, and those with liver biopsies were selected (n = 142). A panel of 65 biomarkers were evaluated by assessing baseline and absolute as well as relative changes at the end of treatment. RESULTS The biomarkers included in E1 score (baseline adiponectin and ferritin; delta of matrix metalloproteinase 9 and transferrin), E2 score (baseline cytokeratin 18 Fragment M65; delta of hyaluronic acid, fructosamine, and alanine aminotransferase), and E3 score (baseline cytokeratin 18 Fragment M65 and gamma-glutamyl transferase; delta of aspartate aminotransferase, insulin, and urea) represented metabolic, apoptotic, and fibrosis aspects of the disease. These signatures provided good accuracy for the noninvasive identification of histologic response under lanifibranor with area under the receiver operating characteristic curve at 0.81 ± 0.08 for E1 score, 0.80 ± 0.08 for E2 score, and 0.81 ± 0.08 for E3 score. CONCLUSIONS Results from this analysis show evidence that baseline values and changes in selected serum biomarkers can aid in predicting histologic response in MASH under lanifibranor treatment. These findings support utilizing a similar approach in a larger sample size (NATiV3, NCT03008070).
Collapse
Affiliation(s)
- Jérôme Boursier
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France; Hepato-Gastroenterology and Digestive Oncology Department, Angers University Hospital, Angers, France.
| | - Hugo Hervé
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| | - Marine Roux
- HIFIH Laboratory, SFR ICAT 4208, Angers University, Angers, France
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Wilrijk, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Ivashkin VT, Drapkina OM, Maevskaya MV, Raikhelson KL, Okovityi SV, Zharkova MS, Grechishnikova VR, Abdulganieva DI, Alekseenko SA, Ardatskaya MD, Bakulin IG, Bakulina NV, Bogomolov PO, Breder VV, Vinnitskaya EV, Geyvandova NI, Golovanova EV, Grinevich VB, Doshchitsin VL, Dudinskaya EN, Ershova EV, Kodzoeva KB, Kozlova IV, Komshilova KA, Konev YV, Korochanskaya NV, Kotovskaya YV, Kravchuk YA, Loranskaya ID, Maev IV, Martynov AI, Mekhtiev SN, Mishina EE, Nadinskaia MY, Nikitin IG, Osipenko MF, Ostroumova OD, Pavlov CS, Pogosova NV, Radchenko VG, Roytberg GE, Saifutdinov RG, Samsonov AA, Seliverstov PV, Sitkin SI, Tarasova LV, Tarzimanova AI, Tkacheva ON, Tkachenko EI, Troshina EA, Turkina SV, Uspenskiy YP, Fominykh YA, Khlynova OV, Tsyganova YV, Shamkhalova MS, Sharkhun OO, Shestakova MV. Clinical Guidelines of the Russian Society for the Study of the Liver, Russian Gastroenterological Association, Russian Society for the Prevention of Non-Communicable Diseases, Russian Association of Endocrinologists, Russian Scientific Medical Society of Therapists, National Society of Preventive Cardiology, Russian Association of Gerontologists and Geriatricians on Non-Alcoholic Fatty Liver Disease. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2025; 35:94-152. [DOI: 10.22416/1382-4376-2025-35-1-94-152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2025]
Abstract
Aim. The clinical guidelines are intended to provide information support for making decisions by gastroenterologists, general practitioners and internists that will improve the quality of medical care for patients with non-alcoholic fatty liver disease, taking into account the latest clinical data and principles of evidence-based medicine. Key points. Clinical guidelines contain information about current views on etiology, risk factors and pathogenesis of nonalcoholic fatty liver disease, peculiarities of its clinical course. Also given recommendations provide information on current methods of laboratory and instrumental diagnostics, invasive and non-invasive tools for nonalcoholic fatty liver disease and its clinical phenotypes assessment, approaches to its treatment, considering the presence of comorbidities, features of dispensary monitoring and prophylaxis. The information is illustrated with algorithms of differential diagnosis and physician's actions. In addition, there is information for the patient and criteria for assessing the quality of medical care. Conclusion. Awareness of specialists in the issues of diagnosis, treatment and follow-up of patients with nonalcoholic fatty liver disease contributes to the timely diagnosis and initiation of treatment, which in the long term will significantly affect their prognosis and quality of life.
Collapse
Affiliation(s)
- V. T. Ivashkin
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| | - M. V. Maevskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - K. L. Raikhelson
- Saint Petersburg State University;
Academician I.P. Pavlov First Saint Petersburg State Medical University
| | - S. V. Okovityi
- Saint Petersburg State Chemical Pharmaceutical University
| | - M. S. Zharkova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | | - M. D. Ardatskaya
- Central State Medical Academy of the Department of Presidential Affairs
| | - I. G. Bakulin
- North-Western State Medical University named after I.I. Mechnikov
| | - N. V. Bakulina
- North-Western State Medical University named after I.I. Mechnikov
| | - P. O. Bogomolov
- Russian University of Medicine;
Moscow Regional Research Clinical Institute
| | - V. V. Breder
- National Medical Research Center of Oncology named after N.N. Blokhin
| | | | | | | | | | | | | | | | - K. B. Kodzoeva
- National Medical Research Center for Transplantology and Artificial Organs named after Academician V.I. Shumakov
| | - I. V. Kozlova
- Saratov State Medical University named after V.I. Razumovsky
| | | | | | | | | | | | | | | | | | - S. N. Mekhtiev
- Academician I.P. Pavlov First Saint Petersburg State Medical University
| | | | - M. Yu. Nadinskaia
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. G. Nikitin
- N.I. Pirogov Russian National Research Medical University;
National Medical Research Center “Treatment and Rehabilitation Center”
| | | | | | - Ch. S. Pavlov
- I.M. Sechenov First Moscow State Medical University (Sechenov University);
Moscow Multidisciplinary Scientific and Clinical Center named after S.P. Botkin
| | - N. V. Pogosova
- National Medical Research Center of Cardiology named after Academician E.I. Chazov
| | | | - G. E. Roytberg
- N.I. Pirogov Russian National Research Medical University
| | - R. G. Saifutdinov
- Kazan State Medical Academy — Branch Campus of the Russian Medical Academy of Continuous Professional Education
| | | | | | - S. I. Sitkin
- North-Western State Medical University named after I.I. Mechnikov;
V.A. Almazov National Medical Research Center
| | | | - A. I. Tarzimanova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - O. N. Tkacheva
- N.I. Pirogov Russian National Research Medical University
| | | | | | | | - Yu. P. Uspenskiy
- Academician I.P. Pavlov First Saint Petersburg State Medical University;
Saint Petersburg State Pediatric Medical University
| | - Yu. A. Fominykh
- V.A. Almazov National Medical Research Center; Saint Petersburg State Pediatric Medical University
| | - O. V. Khlynova
- Perm State Medical University named after Academician E.A. Wagner
| | | | | | - O. O. Sharkhun
- N.I. Pirogov Russian National Research Medical University
| | | |
Collapse
|
11
|
Ghare S, Warner D, Warner J, Chilton PM, Lee J, Zhang J, Wang M, Hardesty J, Treves R, Gabbard J, Anderson C, Batra L, Sreenivasan C, Kraenzle J, McCulley M, McCoy S, Zhang L, Feng W, Gondim DD, Barve S, Zheng J, Palmer K, McClain C, Kirpich I. Impact of chronic ethanol consumption and SARS-COV-2 on the liver and intestine: A pilot dose-response study in mice. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:587-598. [PMID: 39757351 PMCID: PMC11928281 DOI: 10.1111/acer.15528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND During the coronavirus disease 2019 (COVID-19) pandemic, there was a marked increase in alcohol consumption. COVID-19 superimposed on underlying liver disease notably worsens the outcome of many forms of liver injury. The goal of a current pilot study was to test the dual exposure of alcohol and COVID-19 infection in an experimental animal model of alcohol-associated liver disease (ALD). METHODS After 4 weeks of ethanol (EtOH) feeding, C57BL/6 male mice received SARS-CoV-2 (SARS2-N501YMA30) intranasally at 3 × 102, 1 × 103, 3 × 103, and 1 × 104 plaque-forming units (PFU). Mice were then weighed/monitored daily for morbidity/mortality for 10 days while continuing EtOH consumption. Markers of liver inflammation, injury, and intestinal barrier integrity were evaluated. RESULTS A similar gradual weight loss was observed in all inoculated mice (slightly less in the 3 × 102 group) up to post-infection day 4. Greater mortality was observed in mice receiving the highest viral dose at days 3 and 4 post-infection. The majority of the surviving mice subjected to EtOH and inoculated with 3 × 103 or 1 × 104 PFU rapidly lost 25% of their body weight and were euthanized on post-infection day 4. Analysis of liver health in animals that survived to the end of the experiment exhibited no significant changes in hepatic steatosis but had a limited increase in plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels at all viral doses versus EtOH alone. However, the 1 × 104 PFU viral dose exacerbated EtOH-induced hepatic inflammation characterized by elevated levels of several pro-inflammatory cytokines, including Il-6 and Tnf-α. There was limited effect of viral infection on the intestine. CONCLUSIONS SARS-CoV-2 infection caused a dose-dependent negative impact on body weight and survival in mice fed EtOH. This pilot study suggests that early mortality observed after high-dose SARS-CoV-2 challenge could be due, in part, to hepatic dysfunction following chronic EtOH feeding.
Collapse
Affiliation(s)
- Smita Ghare
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Dennis Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jeffrey Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Paula M. Chilton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jiyeon Lee
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - JingWen Zhang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Min Wang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Josiah Hardesty
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Rui Treves
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jon Gabbard
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Charles Anderson
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Lalit Batra
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Chithra Sreenivasan
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jennifer Kraenzle
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Matthew McCulley
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Stephanie McCoy
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Lihua Zhang
- Department of Structural & Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, United States
| | - Wenke Feng
- Department of Structural & Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, United States
| | - Dibson Dibe Gondim
- Department of Pathology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Shirish Barve
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Alcohol Research Center, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Jian Zheng
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Kenneth Palmer
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| | - Craig McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Alcohol Research Center, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Robley Rex Veterans Affairs Medical Center, 800 Zorn Avenue, Louisville, KY 40206, United States
| | - Irina Kirpich
- Alcohol Research Center, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, 505 South Hancock Street, Louisville KY, 40202, United States
| |
Collapse
|
12
|
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
|
13
|
Fujiwara Y, Kuroda H, Abe T, Nagasawa T, Nakaya I, Ito A, Watanabe T, Yusa K, Sato H, Suzuki A, Endo K, Yoshida Y, Oikawa T, Kakisaka K, Sawara K, Tada T, Miyasaka A, Oguri T, Kamiyama N, Matsumoto T. Impact of shear wave elastography and attenuation imaging for predicting life-threatening event in patients with metabolic dysfunction-associated steatotic liver disease. Sci Rep 2025; 15:4547. [PMID: 39915518 PMCID: PMC11802924 DOI: 10.1038/s41598-025-87974-w] [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: 09/27/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
We aimed to elucidate the value of ultrasound-based biomarkers for predicting the major life-threatening events in metabolic dysfunction-associated steatotic liver disease (MASLD). We established a prospective cohort of 279 patients who underwent two-dimensional shear wave elastography (2D-SWE), ultrasound-guided attenuation parameter (UGAP). An area under the curve analysis was performed to determine the cutoff values of liver stiffness measurements (LSM) by 2D-SWE and attenuation coefficient (AC) by UGAP for a moderate fibrosis and a moderate steatosis. We then classified the cohort into Groups A (low LSM and low AC), B (low LSM and high AC), C (high LSM and high AC), and D (high LSM and low AC). We compared the incidence of events between the groups, and estimated the hazard ratios (HRs) with 95% confidence intervals (CIs). The LSM and AC cut off values were 8.37 kPa and 0.62 dB/cm/MHz, respectively. The cumulative incidence rate in Groups A, B, C, and D were 11.2%, 12.2%, 29.5%, and 31.0%/5years, respectively (p < 0.05). LSM (HRs = 1.20, 95%CIs: 1.09-1.32, p < 0.01), and AC (HRs = 1.62, 95%CIs: 1.04-2.51, p = 0.03) were associated with life-threatening events. A combination of 2D-SWE and UGAP may help identify patients with MASLD at high risk for subsequent life-threatening events.
Collapse
Affiliation(s)
- Yudai Fujiwara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan.
| | - Hidekatsu Kuroda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Tamami Abe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Tomoaki Nagasawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Ippeki Nakaya
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Asami Ito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Takuya Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Kenji Yusa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Hiroki Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Akiko Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Kei Endo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Yuichi Yoshida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Takayoshi Oikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Keisuke Kakisaka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Kei Sawara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Toshifumi Tada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akio Miyasaka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Takuma Oguri
- Ultrasound General Imaging, GE HealthCare, Hino, Tokyo, Japan
| | | | - Takayuki Matsumoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| |
Collapse
|
14
|
Pitrone P, Cacciola A, Cattafi A, Romeo AM, Cracò A, Aricò FM, Migliaccio N, Magnani F, Bellone IG, Caloggero S, Mastroeni G. Macro-vacuolar steatosis in a cirrhotic liver mimicking metastatic disease. Radiol Case Rep 2025; 20:1208-1210. [PMID: 39697262 PMCID: PMC11652910 DOI: 10.1016/j.radcr.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/21/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Multinodular steatosis represents a relatively uncommon manifestation of fatty liver disease (FLD). Co-morbidities such as metabolic syndrome or cirrhosis are often associated. Despite typical features of imaging (ultrasound, CT, and MRI), core biopsy sometimes remains the gold standard for diagnosis. We describe the case of a 57-year-old male patient with a long history of hepatic cirrhosis and a recent diagnosis of carcinoma of the tongue, successfully treated. Due to the occurrence of nausea, diarrhea and jaundice the patient is admitted to Our Hospital where ultrasound examination and contrast-enhanced CT are performed, showing global hypoechogenicity of the liver parenchyma with multiple hypo-attenuating lesions. To rule out metastatic lesions, contrast-enhanced CT of the thorax and cranium and gastroscopy and colonoscopy are performed, with no evidence of primary malignancy. Core biopsy reveals macro-vacuolar steatosis within a cirrhotic liver with regenerative aspects.
Collapse
Affiliation(s)
- Pietro Pitrone
- Radiology Unit, "Papardo" Hospital, Messina, ME 98158, Italy
| | - Agatino Cacciola
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", Messina, ME 98124, Italy
| | | | - Alessia Maria Romeo
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", Messina, ME 98124, Italy
| | - Annalisa Cracò
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", Messina, ME 98124, Italy
| | - Francesco Marcello Aricò
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", Messina, ME 98124, Italy
| | - Nicola Migliaccio
- Diagnostic and Interventional Radiology Unit, BIOMORF Department, University Hospital "Policlinico G. Martino", Messina, ME 98124, Italy
| | | | | | | | | |
Collapse
|
15
|
Vali Y, van Dijk A, Lee J, Boursier J, Ratziu V, Yunis C, Schattenberg JM, Valenti L, Gomez MR, Schuppan D, Petta S, Allison M, Hartman ML, Porthan K, Dufour J, Bugianesi E, Gastadelli A, Derdak Z, Fournier‐Poizat C, Shumbayawonda E, Kalutkiewicz M, Yki‐Jarvinen H, Ekstedt M, Geier A, Trylesinski A, Francque S, Brass C, Pavlides M, Holleboom AG, Nieuwdorp M, Anstee QM, Bossuyt PM, the LITMUS investigators. Precision in Liver Diagnosis: Varied Accuracy Across Subgroups and the Need for Variable Thresholds in Diagnosis of MASLD. Liver Int 2025; 45:e16240. [PMID: 39865358 PMCID: PMC11771619 DOI: 10.1111/liv.16240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/18/2024] [Accepted: 12/28/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND AND AIMS The performance of non-invasive liver tests (NITs) is known to vary across settings and subgroups. We systematically evaluated whether the performance of three NITs in detecting advanced fibrosis in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) varies with age, sex, body mass index (BMI), type 2 diabetes mellitus (T2DM) status or liver enzymes. METHODS Data from 586 adult LITMUS Metacohort participants with histologically characterised MASLD were included. The diagnostic performance of the Fibrosis-4 Index (FIB-4), enhanced liver fibrosis (ELF) and vibration-controlled transient elastography liver stiffness measurement (VCTE LSM) was evaluated. Performance was expressed as the area under the receiver operating characteristics curve (AUC). Thresholds for detecting advanced fibrosis (≥F3) were calculated for each NIT for fixed (high) sensitivity, specificity and predictive values. RESULTS Differences in AUC between all subgroups were small and statistically not significant, indicating comparable performance in detecting ≥F3, irrespective of these clinical factors. However, different thresholds were needed to achieve the same level of accuracy with each test. For example, for a fixed sensitivity and specificity, the thresholds for all three NITs were higher in patients with T2DM. Effects for sex, age and liver enzymes were less pronounced. CONCLUSIONS Performance of the selected NITs in detecting advanced liver fibrosis does not vary substantially with clinical characteristics. However, different thresholds have to be selected to achieve the same sensitivity, specificity and predictive values in the respective subgroups. Large prospective studies are called for to study NIT accuracy considering multiple patient characteristics.
Collapse
Affiliation(s)
- Yasaman Vali
- Department of Epidemiology and Data ScienceAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Anne‐Marieke van Dijk
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Internal and Vascular MedicineAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Jenny Lee
- Department of Epidemiology and Data ScienceAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Jerome Boursier
- Laboratoire HIFIH, UPRES EA 3859, SFR ICAT 4208Université d'AngersAngersFrance
- Service d'Hépato‐Gastroentérologie et Oncologie DigestiveCentre Hospitalier Universitaire d'AngersAngersFrance
| | - Vlad Ratziu
- Assistance Publique‐Hôpitaux de Paris, Hôpital Pitié SalpêtrièreICAN (Institute of Cardiometabolism and Nutrition), Sorbonne UniversityParisFrance
| | - Carla Yunis
- Pfizer Research and Development, Pfizer IncLake MaryFloridaUSA
| | - Jörn M. Schattenberg
- Department of Internal Medine IISaarland University Medical CenterHomburgGermany
- Saarland UniversitySaarbrückenGermany
| | - Luca Valenti
- Department of Pathophysiology and TransplantationUniversità Degli Studi di MilanoMilanoItaly
- Precision MedicineBiological Resource Center Unit, Fondazione IRCCS Ca' Granda PoliclinicoMilanoItaly
| | - Manuel Romero Gomez
- Digestive Diseases UnitHospital Universitario Virgen del RocíoSevillaSpain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd)Instituto de Biomedicina de SevillaSevillaSpain
- Universidad de SevillaSevillaSpain
| | - Detlef Schuppan
- Department of Internal Medine IISaarland University Medical CenterHomburgGermany
- Institute of Translational ImmunologyUniversity Medical Center MainzMainzGermany
- Division of GastroenterologyBeth Israel Deaconess Medical CenterBostonMassachusettsUSA
| | - Salvatore Petta
- Sezione di Gastroenterologia e Epatologia, PROMONISE DepartmentUniversità di PalermoPalermoItaly
| | - Mike Allison
- Liver Unit, Department of Medicine, Cambridge NIHR Biomedical Research CentreCambridge University NHS Foundation TrustCambridgeUK
| | - Mark L. Hartman
- Lilly Research LaboratoriesEli Lilly and CompanyIndianapolisIndianaUSA
| | - Kimmo Porthan
- Department of MedicineUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | | | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastro‐Hepatology, A.O. Città della Salute e della Scienza di TorinoUniversity of TurinTurinItaly
| | | | - Zoltan Derdak
- GI DDU, Takeda Pharmaceuticals Company Ltd.CambridgeMassachusettsUSA
| | | | | | | | - Hannele Yki‐Jarvinen
- Department of MedicineUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Mattias Ekstedt
- Department of Health, Medicine and Caring SciencesLinköping UniversityLinköpingSweden
| | - Andreas Geier
- Division of Hepatology, Department Medicine IIWurzburg University HospitalWurzburgGermany
| | | | - Sven Francque
- Department of Gastroenterology Hepatology, and Laboratory of Experimental Medicine and Paediatrics, Antwerp University HospitalUniversity of AntwerpAntwerpBelgium
| | - Clifford Brass
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| | - Michael Pavlides
- Radcliffe Department of Medicine and Oxford NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Adriaan G. Holleboom
- Department of Internal and Vascular MedicineAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular MedicineAmsterdam University Medical CentresAmsterdamThe Netherlands
| | - Quentin M. Anstee
- Translational and Clinical Research Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
- Newcastle NIHR Biomedical Research CentreNewcastle upon Tyne Hospitals NHS TrustNewcastle upon TyneUK
| | - Patrick M. Bossuyt
- Department of Epidemiology and Data ScienceAmsterdam University Medical CentresAmsterdamThe Netherlands
| | | |
Collapse
|
16
|
Chen X, Wei W, Yang F, Wang J, Lv Q, Liu Y, Zhang Z. Bacillus coagulans alleviates hepatic injury caused by Klebsiella pneumoniae in rabbits. PLoS One 2025; 20:e0317252. [PMID: 39792896 PMCID: PMC11723646 DOI: 10.1371/journal.pone.0317252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND As an opportunistic bacterial pathogen, Klebsiella pneumoniae (KP) is prone to causing a spectrum of diseases in rabbits when their immune system is compromised, which poses a threat to rabbit breeding industry. Bacillus coagulans (BC), recognized as an effective probiotic, confers a variety of benefits including anti-inflammatory and antioxidant properties. AIM The purpose of this study was to investigate whether dietary BC can effectively alleviate hepatic injury caused by KP. METHODS In this study, the rabbits were initially pretreated with varying doses of BC (1×106, 5×106, and 1×107 CFU/g), followed by a challenge with KP at a concentration of 1011 CFU/mL. Liver tissues were harvested and processed for histological assessment using H&E and VG stains to assess structural alterations. Biochemical assays were employed to quantify the enzymatic activities of T-SOD and GSH-Px, as well as the MDA content. Furthermore, ELISA was utilized to detect the levels of inflammatory cytokine (IL-10, IL-6, IL-1β and TNF-α) and apoptotic-related gene (Bcl-2, Bax). RESULTS Morphological observation indicated that BC can effectively mitigate KP-induced hepatic sinusoidal dilatation and congestion, as well as ameliorate the degree of hepatic fibrosis. Further analysis showed that BC significantly lowered MDA level in KP-treated rabbits, while enhanced the activities of T-SOD and GSH-Px. Additionally, ELISA result showed that BC pretreatment significantly reduced the levels of pro-inflammatory cytokines TNF-a, IL-6, IL-1β and pro-apoptotic gene Bax, while increasing the levels of anti-inflammatory cytokine IL-10 and anti-apoptotic gene Bcl-2 in KP-treated rabbits. CONCLUSION Above data indicate that BC supplementation effectively attenuated oxidative stress and inflammatory response induced by KP through augmenting the activities of antioxidant enzymes and diminishing the levels of pro-inflammatory factors. Furthermore, it reduced the Bax/Bcl-2 ratio in the liver, thereby inhibiting KP-induced apoptosis. The treatment group receiving 5x106 CFU/g BC benefitted most from the protective effect.
Collapse
Affiliation(s)
- Xiaoguang Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Wenjuan Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Fan Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Jianing Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Qiongxia Lv
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
17
|
Chianelli M, Armellini M, Carpentieri M, Coccaro C, Cuttica CM, Fusco A, Marucci S, Nelva A, Nizzoli M, Ponziani MC, Sciaraffia M, Tassone F, Busetto L. Obesity in Prediabetic Patients: Management of Metabolic Complications and Strategies for Prevention of Overt Diabetes. Endocr Metab Immune Disord Drug Targets 2025; 25:8-36. [PMID: 38778593 PMCID: PMC11826913 DOI: 10.2174/0118715303282327240507184902] [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: 01/07/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/25/2024]
Abstract
Obesity and prediabetes affect a substantial part of the general population, but are largely underdiagnosed, underestimated, and undertreated. Prediabetes differs from diabetes only in the degree of hyperglycaemia consequent to the progressive decline in residual beta-cell function. Both prediabetes and diabetes occur as a consequence of insulin resistance that starts several years before the clinical onset of overt diabetes. Macrovascular complications in patients with diabetes are mainly caused by insulin resistance. This is why in prediabetes, the overall cardiovascular risk is, by all means, similar to that in patients with diabetes. It is important, therefore, to identify prediabetes and treat patients not only to prevent or delay the onset of diabetes, but to reduce the cardiovascular risk associated with prediabetes. This review provides an overview of the pathophysiology of prediabetes in patients with obesity and the progression toward overt diabetes. We have reviewed nutritional and pharmacological approaches to the management of obesity and reduced glucose tolerance, and the treatment of the major comorbidities in these patients, including hypertension, dyslipidaemia, and Metabolic dysfunction-associated Steatotic Liver Disease (MASLD), has also been reviewed. In patients with obesity and prediabetes, the nutritional approach is similar to that adopted for patients with obesity and diabetes; treatments of dyslipidaemia and hypertension also have the same targets compared to patients with diabetes. MASLD is a critical issue in these patients; in the prediabetic state, MASLD rarely progresses into fibrosis. This highlights the importance of the early recognition of this pathological condition before patients become diabetic when the risk of fibrosis is much higher. It is necessary to raise awareness of the clinical relevance of this pathological condition in order to prompt early intervention before complications occur. The single most important therapeutic goal is weight loss, which must be early and persistent.
Collapse
Affiliation(s)
- Marco Chianelli
- Unit of Endocrinology and Metabolism, Regina Apostolorum Hospital, Albano, Rome, Italy
| | - Marina Armellini
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia, Udine, Italy
| | - Maria Carpentieri
- Endocrinology and Metabolism Unit, University-Hospital S. Maria della Misericordia, Udine, Italy
| | - Carmela Coccaro
- Department of Civil Disability, Istituto Nazionale della Previdenza Sociale, Rome, Italy
| | | | - Alessandra Fusco
- Diabetology Center Villaricca, Azienza Sanitaria 2 Naples, Naples, Italy
| | - Simonetta Marucci
- Scienza dell'Alimentazione e Nutrizione Umana, University Campus Biomedico, Rome, Italy
| | - Anna Nelva
- Unit of Endocrinology and Diabetology, Ospedale degli Infermi, Ponderano, Italy
| | - Maurizio Nizzoli
- Unit of Endocrinology and Metabolism G.B. Morgagni Hospital, Forlì, Italy
| | | | | | - Francesco Tassone
- Department of Endocrinology, Diabetes & Metabolism, Santa Croce e Carle Hospital, Cuneo, Italy
| | - Luca Busetto
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
18
|
Ishiba H, Fujii H, Kamada Y, Sumida Y, Takahashi H, Seko Y, Toyoda H, Hayashi H, Yamaguchi K, Iwaki M, Yoneda M, Arai T, Shima T, Morishita A, Kawata K, Tomita K, Kawanaka M, Yoshida Y, Ikegami T, Notsumata K, Oeda S, Fukushima H, Miyoshi E, Aishima S, Itoh Y, Okanoue T, Nakajima A. Accuracy of type IV collagen 7S versus Enhanced Liver Fibrosis score for diagnosing fibrosis in patients with metabolic dysfunction-associated steatotic liver disease. Hepatol Commun 2025; 9:e0563. [PMID: 39670882 PMCID: PMC11637746 DOI: 10.1097/hc9.0000000000000563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Various noninvasive tests can be used to identify high-risk groups of patients with metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD). In this study, we compared the diagnostic performance of serum type 4 collagen 7S (COL4-7S) and the Enhanced Liver Fibrosis (ELF) score for detecting fibrosis in patients with MASLD. METHODS Among 1368 patients with MASLD who underwent liver biopsy, 794 with values for both serum COL4-7S and the ELF score were enrolled in this multicenter study. The diagnostic performance of COL4-7S and ELF for detecting fibrosis stage ≥2, fibrosis stage ≥3, and at-risk metabolic dysfunction-associated steatohepatitis were evaluated using ROC curve, continuous net reclassification improvement, and integrated discrimination improvement analyses. RESULTS Both COL4-7S and ELF scores increased significantly with increasing fibrosis. The AUROC for each outcome was higher for COL4-7S than ELF, but not significantly. The diagnostic performance for detecting fibrosis stage ≥2 was significantly better for COL4-7S than for the ELF score (s net reclassification improvement=16.7%, p=0.018; integrated discrimination improvement=3.9%, p<0.01). In patients without diabetes, the diagnostic performance for each outcome did not differ significantly between COL4-7S and ELF score, but in patients with diabetes, the diagnostic performance for fibrosis stage ≥2 was higher for COL4-7S than for the ELF score (AUROC=0.817 vs. 0.773, p=0.04; s net reclassification improvement=32.7%, p<0.01; integrated discrimination improvement=5.6%, p<0.01). CONCLUSIONS The diagnostic performance of serum COL4-7S (a single marker) for identifying more advanced disease in patients with MASLD was at least equivalent to that of the ELF score (a combined marker).
Collapse
Affiliation(s)
- Hiroshi Ishiba
- Department of Gastroenterology, Osaka General Hospital of West Japan Railway Company, Abeno, Osaka, Japan
| | - Hideki Fujii
- Department of Hepatology, Osaka Metropolitan University, Osaka, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita,Japan
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare University, Tokyo, Japan
| | | | - Yuya Seko
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Hideki Hayashi
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
| | - Kanji Yamaguchi
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | - Taeang Arai
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Toshihide Shima
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kazuhito Kawata
- Department of Internal Medicine II, Hepatology Division, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Kengo Tomita
- Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, General Medical Center, Kawasaki Medical School, Kitaku, Okayama, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Kishibeshinmachi, Osaka, Japan
| | - Tadashi Ikegami
- Department of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Kazuo Notsumata
- Department of General Internal Medicine, Fukui-ken Saiseikai Hospital, 7-1 Wadanaka-chou Funabashi, Fukui, Japan
| | - Satoshi Oeda
- Liver Center, Saga University Hospital, Saga, Japan
| | - Hideaki Fukushima
- Diagnostics Business Area, Siemens Healthcare Diagnostics K.K., Osaki Shinagawa-ku, Tokyo, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry & Clinical Investigation, Osaka University Graduate School of Medicine, Yamada-oka, Suita, Osaka, Japan
| | - Shinichi Aishima
- Department of Scientific Pathology Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| |
Collapse
|
19
|
Naoumov NV, Kleiner DE, Chng E, Brees D, Saravanan C, Ren Y, Tai D, Sanyal AJ. Digital quantitation of bridging fibrosis and septa reveals changes in natural history and treatment not seen with conventional histology. Liver Int 2024; 44:3214-3228. [PMID: 39248039 PMCID: PMC11586893 DOI: 10.1111/liv.16092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/31/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatohepatitis (MASH) with bridging fibrosis is a critical stage in the evolution of fatty liver disease. Second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) microscopy with artificial intelligence (AI) provides sensitive and reproducible quantitation of liver fibrosis. This methodology was applied to gain an in-depth understanding of intra-stage fibrosis changes and septa analyses in a homogenous, well-characterised group with MASH F3 fibrosis. METHODS Paired liver biopsies (baseline [BL] and end of treatment [EOT]) of 57 patients (placebo, n = 17 and tropifexor n = 40), with F3 fibrosis stage at BL according to the clinical research network (CRN) scoring, were included. Unstained sections were examined using SHG/TPEF microscopy with AI. Changes in liver fibrosis overall and in five areas of liver lobules were quantitatively assessed by qFibrosis. Progressive, regressive septa, and 12 septa parameters were quantitatively analysed. RESULTS qFibrosis demonstrated fibrosis progression or regression in 14/17 (82%) patients receiving placebo, while the CRN scoring categorised 11/17 (65%) as 'no change'. Radar maps with qFibrosis readouts visualised quantitative fibrosis dynamics in different areas of liver lobules even in cases categorised as 'No Change'. Measurement of septa parameters objectively differentiated regressive and progressive septa (p < .001). Quantitative changes in individual septa parameters (BL to EOT) were observed both in the 'no change' and the 'regression' subgroups, as defined by the CRN scoring. CONCLUSION SHG/TPEF microscopy with AI provides greater granularity and precision in assessing fibrosis dynamics in patients with bridging fibrosis, thus advancing knowledge development of fibrosis evolution in natural history and in clinical trials.
Collapse
Affiliation(s)
| | - David E. Kleiner
- Laboratory of Pathology, Post‐Mortem SectionNational Cancer InstituteBethesdaMarylandUSA
| | | | | | | | - Yayun Ren
- Histoindex Pte. Ltd.SingaporeSingapore
| | - Dean Tai
- Histoindex Pte. Ltd.SingaporeSingapore
| | - Arun J. Sanyal
- Stravitz‐Sanyal Institute of Liver Disease and Metabolic HealthVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| |
Collapse
|
20
|
Chen J, Lu RS, Diaz-Canestro C, Song E, Jia X, Liu Y, Wang C, Cheung CK, Panagiotou G, Xu A. Distinct changes in serum metabolites and lipid species in the onset and progression of NAFLD in Obese Chinese. Comput Struct Biotechnol J 2024; 23:791-800. [PMID: 38318437 PMCID: PMC10839226 DOI: 10.1016/j.csbj.2024.01.007] [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: 08/28/2023] [Revised: 01/08/2024] [Accepted: 01/14/2024] [Indexed: 02/07/2024] Open
Abstract
INTRODUCTION Metabolic disturbances are major contributors to the onset and progression of non-alcoholic fatty liver disease (NAFLD), which includes a histological spectrum ranging from single steatosis (SS) to non-alcoholic steatohepatitis (NASH). This study aimed to identify serum metabolites and lipids enriched in different histological stages of NAFLD and to explore metabolites/lipids as non-invasive biomarkers in risk prediction of NAFLD and NASH in obese Chinese. METHODS Serum samples and liver biopsies were obtained from 250 NAFLD subjects. Untargeted metabolomic and lipidomic profiling were performed using Liquid Chromatography-Mass Spectrometry. Significantly altered metabolites and lipids were identified by MaAsLin2. Pathway enrichment was conducted with MetaboAnalyst and LIPEA. WGCNA was implemented to construct the co-expression network. Logistic regression models were developed to classify different histological stages of NAFLD. RESULTS A total of 263 metabolites and 550 lipid species were detected in serum samples. Differential analysis and pathway enrichment analysis revealed the progressive patterns in metabolic mechanisms during the transition from normal liver to SS and to NASH, including N-palmitoyltaurine, tridecylic acid, and branched-chain amino acid signaling pathways. The co-expression network showed a distinct correlation between different triglyceride and phosphatidylcholine species with disease severity. Multiple models classifying NAFLD versus normal liver and NASH versus SS identified important metabolic features associated with significant improvement in disease prediction compared to conventional clinical parameters. CONCLUSION Different histological stages of NAFLD are enriched with distinct sets of metabolites, lipids, and metabolic pathways. Integrated algorithms highlight the important metabolic and lipidomic features for diagnosis and staging of NAFLD in obese individuals.
Collapse
Affiliation(s)
- Jiarui Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Leibniz Insitute for Natural Product Research and Infection Biology, Microbiome Dynamics, Hans Knöll Institute, Jena, Germany
| | - Ronald Siyi Lu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Erfei Song
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xi Jia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Cynthia K.Y. Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Gianni Panagiotou
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Leibniz Insitute for Natural Product Research and Infection Biology, Microbiome Dynamics, Hans Knöll Institute, Jena, Germany
- Friedrich Schiller University, Faculty of Biological Sciences, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Pharmacology and Pharmacy, the University of Hong Kong, Hong Kong Special Administrative Region
| |
Collapse
|
21
|
Makino K, Ishii T, Ogiso S, Nakakura A, Nishio T, Fukumitsu K, Uebayashi EY, Munekage F, Horie H, Iwaki K, Ito T, Hatano E. Combination of risk alleles of PNPLA3, TM6SF2, and HSD17B13 of donors can predict recurrence of steatotic liver disease after liver transplantation. Hepatol Res 2024; 54:1148-1157. [PMID: 39031833 DOI: 10.1111/hepr.14086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/25/2024] [Accepted: 06/05/2024] [Indexed: 07/22/2024]
Abstract
AIMS This study aimed to identify the genetic risk factors from donors or recipients that contribute to postliver transplantation (LT) steatotic liver disease (SLD), focusing on the genetic risk score (GRS) based on single nucleotide polymorphisms (SNPs) in SLD patients. METHODS This retrospective study included 55 Japanese SLD recipients and their respective donors. Genotyping of PNPLA3, TM6SF2, and HSD17B13 was undertaken, and the combined GRS was calculated. The relationship between the GRS and the incidence of posttransplant SLD was also evaluated. RESULTS The SLD recipients had a high prevalence of post-LT graft steatosis/steatohepatitis (76.4% and 58.2%, respectively). Although the recipients had a high frequency of risk alleles, there was no relationship between the number of risk alleles for each SNP and the incidence of posttransplant SLD. In contrast, an increased number of risk alleles for any SNP in the donor was correlated with high incidence rates of both post-LT steatosis and steatohepatitis. A multivariable analysis showed that a high donor GRS was an independent risk factor for graft steatosis (odds ratio 8.77; 95% CI, 1.94-52.94; p = 0.009). Similarly, a high donor GRS was an independent risk factor (odds ratio 6.76; 95% CI, 1.84-30.78; p = 0.007) for post-LT graft steatohepatitis. CONCLUSIONS Donor risk alleles of PNPLA3, TM6SF2, and HSD17B13, rather than recipient risk alleles, have been implicated in the development of posttransplant SLD. The combination of these donor risk alleles into a GRS could predict the development of posttransplant SLD.
Collapse
Affiliation(s)
- Kenta Makino
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takamichi Ishii
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Ogiso
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiyoshi Nakakura
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Nishio
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken Fukumitsu
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Fumiaki Munekage
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Horie
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kentaro Iwaki
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ito
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
22
|
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
|
23
|
Hudson D, Afzaal T, Bualbanat H, AlRamdan R, Howarth N, Parthasarathy P, AlDarwish A, Stephenson E, Almahanna Y, Hussain M, Diaz LA, Arab JP. Modernizing metabolic dysfunction-associated steatotic liver disease diagnostics: the progressive shift from liver biopsy to noninvasive techniques. Therap Adv Gastroenterol 2024; 17:17562848241276334. [PMID: 39553445 PMCID: PMC11565685 DOI: 10.1177/17562848241276334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 07/27/2024] [Indexed: 11/19/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health concern worldwide. Liver biopsy is the gold standard for diagnosing and staging MASLD, but it is invasive and carries associated risks. In recent years, there has been significant progress in developing noninvasive techniques for evaluation. This review article discusses briefly current available noninvasive assessments and the various liver biopsy techniques available for MASLD, including invasive techniques such as transjugular and transcutaneous needle biopsy, intraoperative/laparoscopic biopsy, and the evolving role of endoscopic ultrasound-guided biopsy. In addition to discussing the various biopsy techniques, we review the current state of knowledge on the histopathologic evaluation of MASLD, including the various scoring systems used to grade and stage the disease. We also explore current and alternative modalities for histopathologic evaluation, such as whole slide imaging and the utility of immunohistochemistry. Overall, this review article provides a comprehensive overview of the progress in liver biopsy techniques for MASLD and compares invasive and noninvasive modalities. However, beyond clinical trials, the practical application of liver biopsy may be limited, as ongoing advancements in noninvasive fibrosis assessments are expected to more effectively identify candidates for MASLD treatment in real-world settings.
Collapse
Affiliation(s)
- David Hudson
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Tamoor Afzaal
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Hasan Bualbanat
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Raaed AlRamdan
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Nisha Howarth
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Pavithra Parthasarathy
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Alia AlDarwish
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Emily Stephenson
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Yousef Almahanna
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Maytham Hussain
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Luis Antonio Diaz
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- MASLD Research Center, Division of MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
| | - Juan Pablo Arab
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, 1201 E. Broad St. P.O. Box 980341, Richmond, VA 23284, USA
| |
Collapse
|
24
|
Kaylan KB, Paul S. NAFLD No More: A Review of Current Guidelines in the Diagnosis and Evaluation of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Curr Diab Rep 2024; 25:5. [PMID: 39535566 DOI: 10.1007/s11892-024-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW Provide a concise update on metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), as well as a practical approach to screening and initial evaluation. RECENT FINDINGS Nomenclature changes have placed a greater focus on cardiometabolic risk factors in the definition of MASLD. Screening for MASLD is by stepwise noninvasive serum and imaging tests which can identify patients at risk for advanced fibrosis and liver-related complications. MASLD has been increasing in prevalence and disease burden but is underrecognized in primary care and endocrinology clinics. Multiple society guidelines, synthesized here, provide a framework for the initial approach in the diagnosis and evaluation of MASLD. Recent advances in pharmacologic treatment underline the importance of screening for patients who are at risk for advanced fibrosis as they are most likely to benefit from new drug classes, such as the liver-directed thyroid receptor agonist resmiterom.
Collapse
Affiliation(s)
- Kerim B Kaylan
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Medicine, Chicago, IL, USA
| | - Sonali Paul
- Section of Gastroenterology, Hepatology, and Nutrition, Center for Liver Diseases, The University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
25
|
Soluyanova P, Quintás G, Pérez-Rubio Á, Rienda I, Moro E, van Herwijnen M, Verheijen M, Caiment F, Pérez-Rojas J, Trullenque-Juan R, Pareja E, Jover R. The Development of a Non-Invasive Screening Method Based on Serum microRNAs to Quantify the Percentage of Liver Steatosis. Biomolecules 2024; 14:1423. [PMID: 39595599 PMCID: PMC11592063 DOI: 10.3390/biom14111423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is often asymptomatic and underdiagnosed; consequently, there is a demand for simple, non-invasive diagnostic tools. In this study, we developed a method to quantify liver steatosis based on miRNAs, present in liver and serum, that correlate with liver fat. The miRNAs were analyzed by miRNAseq in liver samples from two cohorts of patients with a precise quantification of liver steatosis. Common miRNAs showing correlation with liver steatosis were validated by RT-qPCR in paired liver and serum samples. Multivariate models were built using partial least squares (PLS) regression to predict the percentage of liver steatosis from serum miRNA levels. Leave-one-out cross validation and external validation were used for model selection and to estimate predictive performance. The miRNAseq results disclosed (a) 144 miRNAs correlating with triglycerides in a set of liver biobank samples (n = 20); and (b) 124 and 102 miRNAs correlating with steatosis by biopsy digital image and MRI analyses, respectively, in liver samples from morbidly obese patients (n = 24). However, only 35 miRNAs were common in both sets of samples. RT-qPCR allowed to validate the correlation of 10 miRNAs in paired liver and serum samples. The development of PLS models to quantitatively predict steatosis demonstrated that the combination of serum miR-145-3p, 122-5p, 143-3p, 500a-5p, and 182-5p provided the lowest root mean square error of cross validation (RMSECV = 1.1, p-value = 0.005). External validation of this model with a cohort of mixed MASLD patients (n = 25) showed a root mean squared error of prediction (RMSEP) of 5.3. In conclusion, it is possible to predict the percentage of hepatic steatosis with a low error rate by quantifying the serum level of five miRNAs using a cost-effective and easy-to-implement RT-qPCR method.
Collapse
Affiliation(s)
- Polina Soluyanova
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Guillermo Quintás
- Health and Biomedicine, LEITAT Technological Center, 08225 Terrassa, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, 28029 Madrid, Spain
| | - Álvaro Pérez-Rubio
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Iván Rienda
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (I.R.); (J.P.-R.)
| | - Erika Moro
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Marcel van Herwijnen
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Marcha Verheijen
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Florian Caiment
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Judith Pérez-Rojas
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (I.R.); (J.P.-R.)
| | - Ramón Trullenque-Juan
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Eugenia Pareja
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Ramiro Jover
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
26
|
Liu K, Feng M, Chi W, Cao Z, Wang X, Ding Y, Zhao G, Li Z, Lin L, Bao S, Wang H. Liver fibrosis is closely linked with metabolic-associated diseases in patients with autoimmune hepatitis. Hepatol Int 2024; 18:1528-1539. [PMID: 39249647 PMCID: PMC11461548 DOI: 10.1007/s12072-024-10727-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/25/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND This cross-sectional study aimed to investigate the impact of metabolic-associated diseases (MADs) on patients with autoimmune hepatitis (AIH). METHODS The study analyzed the clinical characteristics of 283 AIH patients who underwent liver biopsy between January 2016 and February 2022 in Ruijin Hospital, Shanghai, China. RESULTS Among the identified AIH patients (n = 283), 87.3%, 23.0%, or 43.1% had MADs, non-alcoholic fatty liver disease (NAFLD), or severe fibrosis, respectively. The proportion of diabetes mellitus (DM) was significantly higher in patients with severe liver fibrosis than in those with mild or moderate fibrosis in the AIH cohort (31.1% vs. 18.0%, p < 0.05). Fibrosis was also more severe in patients with NAFLD than in those without (53.8% vs. 39.9%, p < 0.05). Age, Plts, IgG and the presence with MADs were identified as independent predictors of the severity of inflammation in AIH patients. Moreover, severe liver fibrosis (stages 3 to 4) was independently associated with male (OR, 2.855; p = 0.025), γ-GT (OR, 0.997; p = 0.007), and combination with MADs (OR, 4.917; p = 0.006). Furthermore, combination with DM was also an independent predictor of severe liver fibrosis in AIH patients (OR, 2.445, p = 0.038). CONCLUSIONS Concurrent MADs, common in AIH patients, is an independent risk factor for severe fibrosis or inflammation; of note, combination with DM was also an independent predictor of severe liver fibrosis in AIH patients. While managing with AIH, routine assessment of co-existing MADs, especially DM, is also important.
Collapse
Affiliation(s)
- Kehui Liu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mingyang Feng
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wanqing Chi
- Epidemiology of Microbial Disease, Yale University School of Public Health, New Haven, USA
| | - Zhujun Cao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyin Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yezhou Ding
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Gangde Zhao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ziqiang Li
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lanyi Lin
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shisan Bao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
27
|
Iyer JS, Juyal D, Le Q, Shanis Z, Pokkalla H, Pouryahya M, Pedawi A, Stanford-Moore SA, Biddle-Snead C, Carrasco-Zevallos O, Lin M, Egger R, Hoffman S, Elliott H, Leidal K, Myers RP, Chung C, Billin AN, Watkins TR, Patterson SD, Resnick M, Wack K, Glickman J, Burt AD, Loomba R, Sanyal AJ, Glass B, Montalto MC, Taylor-Weiner A, Wapinski I, Beck AH. AI-based automation of enrollment criteria and endpoint assessment in clinical trials in liver diseases. Nat Med 2024; 30:2914-2923. [PMID: 39112795 PMCID: PMC11485234 DOI: 10.1038/s41591-024-03172-7] [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: 05/02/2023] [Accepted: 07/03/2024] [Indexed: 09/08/2024]
Abstract
Clinical trials in metabolic dysfunction-associated steatohepatitis (MASH, formerly known as nonalcoholic steatohepatitis) require histologic scoring for assessment of inclusion criteria and endpoints. However, variability in interpretation has impacted clinical trial outcomes. We developed an artificial intelligence-based measurement (AIM) tool for scoring MASH histology (AIM-MASH). AIM-MASH predictions for MASH Clinical Research Network necroinflammation grades and fibrosis stages were reproducible (κ = 1) and aligned with expert pathologist consensus scores (κ = 0.62-0.74). The AIM-MASH versus consensus agreements were comparable to average pathologists for MASH Clinical Research Network scores (82% versus 81%) and fibrosis (97% versus 96%). Continuous scores produced by AIM-MASH for key histological features of MASH correlated with mean pathologist scores and noninvasive biomarkers and strongly predicted progression-free survival in patients with stage 3 (P < 0.0001) and stage 4 (P = 0.03) fibrosis. In a retrospective analysis of the ATLAS trial (NCT03449446), responders receiving study treatment showed a greater continuous change in fibrosis compared with placebo (P = 0.02). Overall, these results suggest that AIM-MASH may assist pathologists in histologic review of MASH clinical trials, reducing inter-rater variability on trial outcomes and offering a more sensitive and reproducible measure of patient responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aryan Pedawi
- PathAI, Boston, MA, USA
- Atomwise, San Francisco, CA, USA
| | | | | | | | - Mary Lin
- PathAI, Boston, MA, USA
- Supernus Pharmaceuticals, Rockville, MD, USA
| | | | - Sara Hoffman
- PathAI, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Hunter Elliott
- PathAI, Boston, MA, USA
- BigHat Biosciences, San Mateo, CA, USA
| | - Kenneth Leidal
- PathAI, Boston, MA, USA
- Genesis Therapeutics, Burlingame, CA, USA
| | - Robert P Myers
- Gilead Sciences, Inc., Foster City, CA, USA
- OrsoBio, Inc., Palo Alto, CA, USA
| | - Chuhan Chung
- Gilead Sciences, Inc., Foster City, CA, USA
- Inipharm, San Diego, CA, USA
| | | | | | | | - Murray Resnick
- PathAI, Boston, MA, USA
- Rhode Island Hospital and The Miriam Hospital, Providence, RI, USA
| | | | - Jon Glickman
- PathAI, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Alastair D Burt
- NIHRB Medical Research Center, Newcastle University, Newcastle, UK
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, San Diego, CA, USA
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, VCU School of Medicine, Richmond, VA, USA
| | | | | | | | - Ilan Wapinski
- PathAI, Boston, MA, USA
- Sanofi Pharmaceuticals, Cambridge, MA, USA
| | | |
Collapse
|
28
|
Fouad Y, Alboraie M, Shiha G. Epidemiology and diagnosis of metabolic dysfunction-associated fatty liver disease. Hepatol Int 2024; 18:827-833. [PMID: 38967907 PMCID: PMC11450050 DOI: 10.1007/s12072-024-10704-3] [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: 02/08/2024] [Accepted: 06/01/2024] [Indexed: 07/06/2024]
Abstract
The most common chronic liver illness worldwide is metabolic dysfunction linked to fatty liver disease (MAFLD), which is poorly understood by doctors and patients. Many people with this disease develop steatohepatitis, cirrhosis and its consequences, as well as extrahepatic manifestations; these conditions are particularly common if they are linked to diabetes mellitus or obesity. A breakthrough with numerous benefits is the switch from NAFLD to MAFLD in terms of terminology and methodology. The diagnosis of MAFLD is based on affirmative criteria; unlike NAFLD, it is no longer based on exclusion. The diagnosis of MAFLD and the evaluation of steatosis and fibrosis is achieved using liver biopsy and non-invasive laboratory or radiographic techniques. We briefly address the most recent developments in MAFLD epidemiology and diagnosis.
Collapse
Affiliation(s)
- Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Minia, Egypt.
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, Egypt
| | - Gamal Shiha
- Egyptian Liver Research Institute and Hospital, Mansoura, Egypt
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
29
|
Saad M, Ibrahim W, Hasanin AH, Elyamany AM, Matboli M. Evaluating the therapeutic potential of genetically engineered probiotic Zbiotics (ZB183) for non-alcoholic steatohepatitis (NASH) management via modulation of the cGAS-STING pathway. RSC Med Chem 2024:d4md00477a. [PMID: 39290381 PMCID: PMC11403872 DOI: 10.1039/d4md00477a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
NAFLD/NASH has emerged as a global health concern with no FDA-approved treatment, necessitating the exploration of novel therapeutic elements for NASH. Probiotics are known as an important adjunct therapy in NASH. Zbiotics (ZB183) is the first commercially available genetically engineered probiotic. Herein, we aimed to evaluate the potential therapeutic effects of Zbiotics administration on NASH management by modulating the cGAS-STING-signaling pathway-related RNA network. In silico data analysis was performed and three DEGs (MAPK3/EDN1/TNF) were selected with their epigenetic modulators (miR-6888-5p miRNA, and lncRNA RABGAP1L-DT-206). The experimental design included NASH induction with an HSHF diet in Wistar rats and Zbiotics administration in NASH rats in comparison to statin treatment. Liver functions and lipid profile were assessed. Additionally, the expression levels of the constructed molecular network were assessed using RT-PCR. Moreover, the Zbiotics effects in NASH were further validated with histopathological examination of liver and colon samples. Also, immunohistochemistry staining of hepatic TNF-α and colonic occludin was assessed. Oral administration of Zbiotics for four weeks downregulated the expression of the cGAS-STING-related network (MAPK3/EDN1/TNF/miR-6888-5p miRNA/lncRNA RABGAP1L-DT-206) in NASH models. Zbiotics also ameliorated hepatic inflammation and steatosis, as evidenced by a notable improvement in NAS score and decreased hepatic TNF-α levels. Furthermore, Zbiotics exhibited favorable effects on colon health, including increased crypt length, reduced inflammatory cell infiltration, and restoration of colonic mucosa occludin expression. In conclusion, our findings suggest that Zbiotics has potential therapeutic effects on NASH via modulating the gut-liver axis and the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Maha Saad
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Modern University for Technology and Information Cairo Egypt
- Biomedical Research Department, Faculty of Medicine, Modern University for technology and information Cairo Egypt
- Medical Biochemistry and Molecular Biology, Faculty of Medicine Cairo University Cairo Egypt
| | - Walaa Ibrahim
- Medical Biochemistry and Molecular Biology, Faculty of Medicine Cairo University Cairo Egypt
| | - Amany Helmy Hasanin
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University Cairo 11566 Egypt
| | - Aya Magdy Elyamany
- Anatomic Pathology Department, Faculty of Medicine, Cairo University Cairo Egypt
| | - Marwa Matboli
- Departement of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University Cairo 11566 Egypt
| |
Collapse
|
30
|
Gargiulo S, Barone V, Bonente D, Tamborrino T, Inzalaco G, Gherardini L, Bertelli E, Chiariello M. Integrated Ultrasound Characterization of the Diet-Induced Obesity (DIO) Model in Young Adult c57bl/6j Mice: Assessment of Cardiovascular, Renal and Hepatic Changes. J Imaging 2024; 10:217. [PMID: 39330437 PMCID: PMC11433005 DOI: 10.3390/jimaging10090217] [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: 07/09/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Consuming an unbalanced diet and being overweight represent a global health problem in young people and adults of both sexes, and may lead to metabolic syndrome. The diet-induced obesity (DIO) model in the C57BL/6J mouse substrain that mimics the gradual weight gain in humans consuming a "Western-type" (WD) diet is of great interest. This study aims to characterize this animal model, using high-frequency ultrasound imaging (HFUS) as a complementary tool to longitudinally monitor changes in the liver, heart and kidney. Long-term WD feeding increased mice body weight (BW), liver/BW ratio and body condition score (BCS), transaminases, glucose and insulin, and caused dyslipidemia and insulin resistance. Echocardiography revealed subtle cardiac remodeling in WD-fed mice, highlighting a significant age-diet interaction for some left ventricular morphofunctional parameters. Qualitative and parametric HFUS analyses of the liver in WD-fed mice showed a progressive increase in echogenicity and echotexture heterogeneity, and equal or higher brightness of the renal cortex. Furthermore, renal circulation was impaired in WD-fed female mice. The ultrasound and histopathological findings were concordant. Overall, HFUS can improve the translational value of preclinical DIO models through an integrated approach with conventional methods, enabling a comprehensive identification of early stages of diseases in vivo and non-invasively, according to the 3Rs.
Collapse
Affiliation(s)
- Sara Gargiulo
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Denise Bonente
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | | | - Giovanni Inzalaco
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Lisa Gherardini
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Eugenio Bertelli
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Mario Chiariello
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| |
Collapse
|
31
|
Zhou Y, Nie M, Mao F, Zhou H, Zhao L, Ding J, Kan Y, Jing X. Associations Between Multiparametric US-Based Indicators and Pathological Status in Patients with Metabolic Associated Fatty Liver Disease. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1395-1402. [PMID: 38871490 DOI: 10.1016/j.ultrasmedbio.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/05/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE Noninvasive evaluation of metabolic dysfunction-associated fatty liver disease (MAFLD) using ultrasonography holds significant clinical value. The associations between ultrasound (US)-based parameters and the pathological spectra remain unclear and controversial. This study aims to investigate the associations thoroughly. METHODS The participants with MAFLD undergoing liver biopsy and multiparametric ultrasonography were prospectively recruited from December 2020 to September 2022. Three US-based parameters, namely attenuation coefficient (AC), liver stiffness (LS) and dispersion slope (DS) were obtained. The relationship between these parameters and steatosis grades, inflammation grades and fibrosis stages was examined. RESULTS In this study with 116 participants, AC values significantly differed across distinct steatosis grades (p < 0.001), while DS and LS values varied among inflammation grades (p < 0.001) and fibrosis stages (p < 0.001). The area under the receiver operating characteristic curves (AUCs) of AC ranged from 0.82 to 0.84 for differentiating steatosis grades, while AUCs of LS ranged from 0.62 to 0.76 for distinguishing inflammation grades and 0.83-0.95 for discerning fibrosis stages. AUCs for DS ranged from 0.79 to 0.81 in discriminating inflammation grades and 0.80-0.88 for differentiating fibrosis stages. Subgroup analysis revealed that LS demonstrated different trends in inflammation grade but consistent trends in fibrosis stage across subgroups, whereas DS showed consistent trends for both inflammation grade and fibrosis stage across all subgroups. CONCLUSION AC values indicate the degree of hepatic steatosis but not inflammation or fibrosis. LS values are determined only by fibrosis stage and are not associated with inflammation grades. DS values are associated with both fibrosis and inflammation grades.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal, Life Support for Critical Diseases, Artificial Cell Engineering Technology Research, Center, Tianjin Third Central Hospital, Tianjin, China
| | - Mengjin Nie
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Feng Mao
- Department of Ultrasound, Zhongshan Hospital Fudan University, Shanghai
| | - Hongyu Zhou
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal, Life Support for Critical Diseases, Artificial Cell Engineering Technology Research, Center, Tianjin Third Central Hospital, Tianjin, China
| | - Lin Zhao
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal, Life Support for Critical Diseases, Artificial Cell Engineering Technology Research, Center, Tianjin Third Central Hospital, Tianjin, China
| | - Jianmin Ding
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal, Life Support for Critical Diseases, Artificial Cell Engineering Technology Research, Center, Tianjin Third Central Hospital, Tianjin, China
| | - Yanmin Kan
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal, Life Support for Critical Diseases, Artificial Cell Engineering Technology Research, Center, Tianjin Third Central Hospital, Tianjin, China
| | - Xiang Jing
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin Key Laboratory of Extracorporeal, Life Support for Critical Diseases, Artificial Cell Engineering Technology Research, Center, Tianjin Third Central Hospital, Tianjin, China.
| |
Collapse
|
32
|
Sergi CM. NAFLD (MASLD)/NASH (MASH): Does It Bother to Label at All? A Comprehensive Narrative Review. Int J Mol Sci 2024; 25:8462. [PMID: 39126031 PMCID: PMC11313354 DOI: 10.3390/ijms25158462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), or metabolic dysfunction-associated steatotic liver disease (MASLD), is a liver condition that is linked to overweight, obesity, diabetes mellitus, and metabolic syndrome. Nonalcoholic steatohepatitis (NASH), or metabolic dysfunction-associated steatohepatitis (MASH), is a form of NAFLD/MASLD that progresses over time. While steatosis is a prominent histological characteristic and recognizable grossly and microscopically, liver biopsies of individuals with NASH/MASH may exhibit several other abnormalities, such as mononuclear inflammation in the portal and lobular regions, hepatocellular damage characterized by ballooning and programmed cell death (apoptosis), misfolded hepatocytic protein inclusions (Mallory-Denk bodies, MDBs), megamitochondria as hyaline inclusions, and fibrosis. Ballooning hepatocellular damage remains the defining feature of NASH/MASH. The fibrosis pattern is characterized by the initial expression of perisinusoidal fibrosis ("chicken wire") and fibrosis surrounding the central veins. Children may have an alternative form of progressive NAFLD/MASLD characterized by steatosis, inflammation, and fibrosis, mainly in Rappaport zone 1 of the liver acinus. To identify, synthesize, and analyze the scientific knowledge produced regarding the implications of using a score for evaluating NAFLD/MASLD in a comprehensive narrative review. The search for articles was conducted between 1 January 2000 and 31 December 2023, on the PubMed/MEDLINE, Scopus, Web of Science, and Cochrane databases. This search was complemented by a gray search, including internet browsers (e.g., Google) and textbooks. The following research question guided the study: "What are the basic data on using a score for evaluating NAFLD/MASLD?" All stages of the selection process were carried out by the single author. Of the 1783 articles found, 75 were included in the sample for analysis, which was implemented with an additional 25 articles from references and gray literature. The studies analyzed indicated the beneficial effects of scoring liver biopsies. Although similarity between alcoholic steatohepatitis (ASH) and NASH/MASH occurs, some patterns of hepatocellular damage seen in alcoholic disease of the liver do not happen in NASH/MASH, including cholestatic featuring steatohepatitis, alcoholic foamy degeneration, and sclerosing predominant hyaline necrosis. Generally, neutrophilic-rich cellular infiltrates, prominent hyaline inclusions and MDBs, cholestasis, and obvious pericellular sinusoidal fibrosis should favor the diagnosis of alcohol-induced hepatocellular injury over NASH/MASH. Multiple grading and staging methods are available for implementation in investigations and clinical trials, each possessing merits and drawbacks. The systems primarily used are the Brunt, the NASH CRN (NASH Clinical Research Network), and the SAF (steatosis, activity, and fibrosis) systems. Clinical investigations have utilized several approaches to link laboratory and demographic observations with histology findings with optimal platforms for clinical trials of rapidly commercialized drugs. It is promising that machine learning procedures (artificial intelligence) may be critical for developing new platforms to evaluate the benefits of current and future drug formulations.
Collapse
Affiliation(s)
- Consolato M. Sergi
- Department of Laboratory Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada; ; Tel.: +1-613-737-7600 (ext. 2427); Fax: +1-613-738-4837
- Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
33
|
Ferraioli G, Barr RG, Berzigotti A, Sporea I, Wong VWS, Reiberger T, Karlas T, Thiele M, Cardoso AC, Ayonrinde OT, Castera L, Dietrich CF, Iijima H, Lee DH, Kemp W, Oliveira CP, Sarin SK. WFUMB Guidelines/Guidance on Liver Multiparametric Ultrasound. Part 2: Guidance on Liver Fat Quantification. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1088-1098. [PMID: 38658207 DOI: 10.1016/j.ultrasmedbio.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024]
Abstract
The World Federation for Ultrasound in Medicine and Biology (WFUMB) has promoted the development of this document on multiparametric ultrasound. Part 2 is a guidance on the use of the available tools for the quantification of liver fat content with ultrasound. These are attenuation coefficient, backscatter coefficient, and speed of sound. All of them use the raw data of the ultrasound beam to estimate liver fat content. This guidance has the aim of helping the reader in understanding how they work and interpret the results. Confounding factors are discussed and a standardized protocol for measurement acquisition is suggested to mitigate them. The recommendations were based on published studies and experts' opinion but were not formally graded because the body of evidence remained low at the time of drafting this document.
Collapse
Affiliation(s)
- Giovanna Ferraioli
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Richard Gary Barr
- Department of Radiology, Northeastern Ohio Medical University, Youngstown, OH, USA
| | - Annalisa Berzigotti
- Department for Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ioan Sporea
- Department of Internal Medicine II, Division of Gastroenterology and Hepatology, Center for Advanced Research in Gastroenterology and Hepatology, "Victor Babeș" University of Medicine and Pharmacy, Timișoara, Romania
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Medical Data Analytics Centre, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian-Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Thomas Karlas
- Department of Medicine II, Division of Gastroenterology, Leipzig University Medical Center, Leipzig, Germany
| | - Maja Thiele
- Center for Liver Research, Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark; Department for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ana Carolina Cardoso
- Hepatology Division, School of Medicine, Federal University of Rio de Janeiro, Clementino, Fraga Filho Hospital, Rio de Janeiro, RJ, Brazil
| | - Oyekoya Taiwo Ayonrinde
- Department of Gastroenterology and Hepatology, Fiona Stanley Hospital, Murdoch, WA, Australia; Medical School, The University of Western Australia, Crawley, WA, Australia; Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - Laurent Castera
- Université Paris-Cité, Inserm UMR1149, Centre de Recherche sur l'Inflammation, Paris, France; Service d'Hépatologie, Hôpital Beaujon, Assistance-Publique Hôpitaux de Paris, Clichy, France
| | - Christoph Frank Dietrich
- Department Allgemeine Innere Medizin (DAIM), Kliniken Hirslanden Beau Site, Salem and Permancence, Bern, Switzerland
| | - Hiroko Iijima
- Department of Gastroenterology, Division of Hepatobiliary and Pancreatic Disease, Hyogo Medical University, Nishinomiya, Hyogo, Japan; Ultrasound Imaging Center, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Dong Ho Lee
- Department of Radiology, College of Medicine, Seoul National University Hospital, Seoul National University, Seoul, Republic of Korea
| | - William Kemp
- Department of Gastroenterology, Alfred Hospital, Melbourne, Australia; Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Claudia P Oliveira
- Gastroenterology Department, Laboratório de Investigação (LIM07), Hospital das Clínicas de São Paulo, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
34
|
Castera L, Garteiser P, Laouenan C, Vidal-Trécan T, Vallet-Pichard A, Manchon P, Paradis V, Czernichow S, Roulot D, Larger E, Pol S, Bedossa P, Correas JM, Valla D, Gautier JF, Van Beers BE. Prospective head-to-head comparison of non-invasive scores for diagnosis of fibrotic MASH in patients with type 2 diabetes. J Hepatol 2024; 81:195-206. [PMID: 38548067 DOI: 10.1016/j.jhep.2024.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND & AIMS Non-invasive scores have been proposed to identify patients with fibrotic, metabolic dysfunction-associated steatohepatitis (MASH), who are at the highest risk of progression to complications of cirrhosis and may benefit from pharmacologic treatments. However, data in patients with type 2 diabetes (T2DM) are lacking. The aim of this multicenter prospective study was to perform a head-to-head comparison of FAST (FibroScan-aspartate aminotransferase [AST]), MAST (MRI-AST), MEFIB (magnetic resonance elastography [MRE] plus FIB-4), and FNI (fibrotic NASH index) for detecting fibrotic MASH in patients with T2DM. METHODS A total of 330 outpatients with T2DM and biopsy-proven metabolic dysfunction-associated steatotic liver disease (MASLD) from the QUID-NASH study (NCT03634098), who underwent FibroScan, MRI-proton density fat fraction and MRE at the time of liver biopsy were studied. The main outcome was fibrotic MASH, defined as NAS ≥4 (with at least one point for each parameter) and fibrosis stage ≥2 (centrally reviewed). RESULTS All data for score comparisons were available for 245 patients (median age 59 years, 65% male, median BMI 31 kg/m2; fibrotic MASH in 39%). FAST and MAST had similar accuracy (AUROCs 0.81 vs. 0.79, p = 0.41) but outperformed FNI (0.74; p = 0.01) and MEFIB (0.68; p <0.0001). When using original cut-offs, MAST outperformed FAST, MEFIB and FNI when comparing the percentage of correctly classified patients, in whom liver biopsy would be avoided (69% vs. 48%, 46%, 39%, respectively; p <0.001). When using cut-offs specific to our population, FAST outperformed FNI and MAST (56% vs. 40%, and 38%, respectively; p <0.001). CONCLUSION Our findings show that FAST, MAST, MEFIB and FNI are accurate non-invasive tools to identify patients with T2DM and fibrotic MASH in secondary/tertiary diabetes clinics. Cut-offs adapted to the T2DM population should be considered. IMPACT AND IMPLICATIONS Among patients with type 2 diabetes (T2DM), identifying those with metabolic dysfunction-associated steatohepatitis and significant fibrosis, who are the most at risk of developing clinical liver-related outcomes and who may benefit from pharmacologic treatments, is an unmet need. In this prospective multicenter study, we compared four non-invasive scores, three based on imaging (MRI or ultrasound technologies) and one on laboratory blood tests, for this purpose, using original and study-specific cut-offs. Our findings show that FAST, MAST, MEFIB and FNI are accurate non-invasive tools to identify patients with T2DM and fibrotic MASH in secondary/tertiary diabetes clinics. Cut-offs adapted to the T2DM population should be considered. TRIAL REGISTRATION NUMBER NCT03634098.
Collapse
Affiliation(s)
- Laurent Castera
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'hépatologie, AP-HP, Hôpital Beaujon, F-92110 Clichy-la-Garenne, France.
| | | | - Cédric Laouenan
- Université Paris Cité, UMR1137 (IAME), Inserm, F-75018 Paris, France; (DEBRC), APHP, Hôpital Bichat, Paris, France
| | - Tiphaine Vidal-Trécan
- Centre Universitaire du Diabète et de ses Complications, AP-HP, Hôpital Lariboisière, F-75010 Paris, France
| | | | | | - Valérie Paradis
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'anatomie et de cytologie pathologiques, AP-HP, Hôpital Beaujon, 792110 Clichy-la-Garenne, France
| | - Sébastien Czernichow
- INSERM UMR-S1151, CNRS UMR-S8253, Immediab lab, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France; Service de nutrition, centre spécialisé Obésité, APHP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Dominique Roulot
- Université Paris-Est, U955, Inserm, F-94000 Créteil, France; Unité d'hépatologie, AP-HP, Hôpital Avicenne, 93000 Bobigny, France
| | - Etienne Larger
- Université Paris Cité, Institut Cochin, U1016, Inserm, F-75014 Paris, France; Service de diabétologie, AP-HP, Groupe hospitalier Cochin, F-75014 Paris, France
| | - Stanislas Pol
- Service d'hépatologie, AP-HP, Groupe hospitalier Cochin, F-75014 Paris, France; Université Paris Cité, Institut Cochin, U1016, Inserm, F-75014 Paris, France
| | - Pierre Bedossa
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Liverpat, F-75116 Paris, France
| | - Jean-Michel Correas
- Sorbonne Université, CNRS, INSERM Laboratoire d'Imagerie Biomédicale, Paris, France; Service d'Imagerie Adulte, AP-HP, Hôpital Necker Enfants Malades, F-75015 Paris, France
| | - Dominique Valla
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'hépatologie, AP-HP, Hôpital Beaujon, F-92110 Clichy-la-Garenne, France
| | - Jean-François Gautier
- Centre Universitaire du Diabète et de ses Complications, AP-HP, Hôpital Lariboisière, F-75010 Paris, France; INSERM UMR-S1151, CNRS UMR-S8253, Immediab lab, Institut Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Bernard E Van Beers
- Université Paris Cité, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service de Radiologie, AP-HP, Hôpital Beaujon, F-92110 Clichy-la-Garenne, France
| |
Collapse
|
35
|
Umemura A, Sasaki A, Kumagai H, Tanahashi Y, Iwasaki T, Nitta H. Relationships Between Changes in Serum Ketone Body Levels and Metabolic Effects in Patients with Severe Obesity Who Underwent Laparoscopic Sleeve Gastrectomy. Obes Surg 2024; 34:2607-2616. [PMID: 38842760 PMCID: PMC11217106 DOI: 10.1007/s11695-024-07337-8] [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: 04/04/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Serum ketone bodies increase due to dynamic changes in the lipid metabolisms of patients undergoing bariatric surgery. However, there have been few studies on the role of ketone bodies after bariatric surgery. We aimed to clarify the role of and relationship between the changes in serum ketone bodies and weight loss, as well as between those changes and the metabolic effects after laparoscopic sleeve gastrectomy (LSG). METHODS We recruited 52 patients with severe obesity who underwent LSG. We measured acetoacetic acid (AcAc) and β-hydroxybutyric acid (β-OHB) at the baseline, 1 month, and 6 months after LSG. Subsequently, we compared the changes in the serum ketone bodies with weight-loss effects and various metabolic parameters. RESULTS At 1 month after LSG, β-OHB significantly increased (p = 0.009), then significantly decreased 6 months after LSG (p = 0.002). In addition, β-OHB in patients without Type 2 diabetes (T2D) and metabolic dysfunction-associated steatohepatitis (MASH) was notably higher than in patients with T2D at 1 month after LSG (p < 0.001). In the early phase, both AcAc and β-OHB mainly had strong positive correlations with changes in T2D- and MASH-related parameters. In the middle term after LSG, changes in both AcAc and β-OHB were positively correlated with changes in lipid parameters and chronic kidney disease-related parameters. CONCLUSION We demonstrated that the postoperative surge of ketone bodies plays a crucial function in controlling metabolic effects after LSG. These findings suggest the cause- and consequence-related roles of ketone bodies in the metabolic benefits of bariatric surgery.
Collapse
Affiliation(s)
- Akira Umemura
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan.
| | - Akira Sasaki
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Hideki Kumagai
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Yota Tanahashi
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Takafumi Iwasaki
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Hiroyuki Nitta
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| |
Collapse
|
36
|
Ratziu V, Francque S, Behling CA, Cejvanovic V, Cortez-Pinto H, Iyer JS, Krarup N, Le Q, Sejling AS, Tiniakos D, Harrison SA. Artificial intelligence scoring of liver biopsies in a phase II trial of semaglutide in nonalcoholic steatohepatitis. Hepatology 2024; 80:173-185. [PMID: 38112484 PMCID: PMC11185915 DOI: 10.1097/hep.0000000000000723] [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: 06/28/2023] [Accepted: 12/03/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND AND AIMS Artificial intelligence-powered digital pathology offers the potential to quantify histological findings in a reproducible way. This analysis compares the evaluation of histological features of NASH between pathologists and a machine-learning (ML) pathology model. APPROACH AND RESULTS This post hoc analysis included data from a subset of patients (n=251) with biopsy-confirmed NASH and fibrosis stage F1-F3 from a 72-week randomized placebo-controlled trial of once-daily subcutaneous semaglutide 0.1, 0.2, or 0.4 mg (NCT02970942). Biopsies at baseline and week 72 were read by 2 pathologists. Digitized biopsy slides were evaluated by PathAI's NASH ML models to quantify changes in fibrosis, steatosis, inflammation, and hepatocyte ballooning using categorical assessments and continuous scores. Pathologist and ML-derived categorical assessments detected a significantly greater percentage of patients achieving the primary endpoint of NASH resolution without worsening of fibrosis with semaglutide 0.4 mg versus placebo (pathologist 58.5% vs. 22.0%, p < 0.0001; ML 36.9% vs. 11.9%; p =0.0015). Both methods detected a higher but nonsignificant percentage of patients on semaglutide 0.4 mg versus placebo achieving the secondary endpoint of liver fibrosis improvement without NASH worsening. ML continuous scores detected significant treatment-induced responses in histological features, including a quantitative reduction in fibrosis with semaglutide 0.4 mg versus placebo ( p =0.0099) that could not be detected using pathologist or ML categorical assessment. CONCLUSIONS ML categorical assessments reproduced pathologists' results of histological improvement with semaglutide for steatosis and disease activity. ML-based continuous scores demonstrated an antifibrotic effect not measured by conventional histopathology.
Collapse
Affiliation(s)
- Vlad Ratziu
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié Salpêtrière, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Sven Francque
- Antwerp University Hospital, Antwerp, Belgium
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Antwerp, Belgium
| | | | | | - Helena Cortez-Pinto
- Clínica Universitária de Gastrenterologia, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | | | - Quang Le
- PathAI Inc., Boston, Massachusetts, USA
| | | | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Pathology, Aretaieion Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
37
|
Ogawa S, Kumada T, Gotoh T, Niwa F, Toyoda H, Tanaka J, Shimizu M. A comparative study of hepatic steatosis using two different qualitative ultrasound techniques measured based on magnetic resonance imaging-derived proton density fat fraction. Hepatol Res 2024; 54:638-654. [PMID: 38294946 DOI: 10.1111/hepr.14019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
AIM This study aimed to evaluate the diagnostic performance of attenuation measurement (ATT; dual-frequency method) and improved algorithm of ATT (iATT; reference method) for the assessment of hepatic steatosis using magnetic resonance imaging (MRI)-derived proton density fat fraction (PDFF) as the reference standard. METHODS We prospectively analyzed 427 patients with chronic liver disease who underwent ATT, iATT, or MRI-derived PDFF. Correlation coefficients were analyzed, and diagnostic values were evaluated by area under the receiver operating characteristic curve (AUROC). The steatosis grade was categorized as S0 (<5.2%), S1 (≥5.2%, <11.3%), S2 (≥11.3%, <17.1%), and S3 (≥17.1%) according to MRI-derived PDFF values. RESULTS The median ATT and iATT values were 0.61 dB/cm/MHz (interquartile range 0.55-0.67 dB/cm/MHz) and 0.66 dB/cm/MHz (interquartile range 0.57-0.77 dB/cm/MHz). ATT and iATT values increased significantly as the steatosis grade increased in the order S0, S1, S2, and S3 (p < 0.001). The correlation coefficients between ATT or iATT values and MRI-derived PDFF values were 0.533 (95% confidence interval [CI] 0.477-0.610) and 0.803 (95% CI 0.766-0.834), with a significant difference between them (p < 0.001). For the detection of hepatic steatosis of ≥S1, ≥S2, and ≥S3, iATT yielded AUROCs of 0.926 (95% CI 0.901-0.951), 0.913 (95% CI 0.885-0.941), and 0.902 (95% CI 0.869-0.935), with significantly higher AUROC values than for ATT (p < 0.001, p < 0.001, p = 0.001). CONCLUSION iATT showed excellent diagnostic performance for hepatic steatosis, and was strongly correlated with MRI-derived PDFF, with AUROCs of ≥0.900.
Collapse
Affiliation(s)
- Sadanobu Ogawa
- Department of Imaging Diagnosis, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Takashi Kumada
- Department of Nursing, Faculty of Nursing, Gifu Kyoritsu University, Ogaki, Gifu, Japan
| | - Tatsuya Gotoh
- Department of Imaging Diagnosis, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Fumihiko Niwa
- Department of Imaging Diagnosis, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Gifu, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control, and Prevention, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Masahito Shimizu
- Department of Gastroenterology/Internal Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
38
|
Jönsson C, Bergram M, Kechagias S, Nasr P, Ekstedt M. Activin A levels in metabolic dysfunction-associated steatotic liver disease associates with fibrosis and the PNPLA3 I148M variant. Scand J Gastroenterol 2024; 59:737-741. [PMID: 38563432 DOI: 10.1080/00365521.2024.2334804] [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: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver condition worldwide. There is an urgent need to develop new biomarkers to assess disease severity and to define patients with a progressive phenotype. Activin A is a new promising biomarker with conflicting results about liver fibrosis. In this study we investigate levels of Activin A in patients with biopsy proven MASLD. We assess levels of Activin A in regard to fibrosis stage and genetic variant I148M in the patatin-like phospholipase domain-containing protein 3 (PNPLA3). METHODS Activin A levels were assessed in plasma samples from patients with biopsy-proven MASLD in a cross-sectional study. All patients were clinically evaluated and the PNPLA3 I148M genotype of the cohort was assessed. FINDINGS 41 patients were included and 27% of these had advanced fibrosis. In MASLD patients with advanced fibrosis, Activin A levels was higher (p < 0.001) and could classify advanced fibrosis with an AUROC for activin A of 0.836 (p < 0.001). Patients homozygous for PNPLA3 I148M G/G had higher levels of activin A than non-homozygotes (p = 0.027). CONCLUSIONS Circulating activin A levels were associated with advanced fibrosis and could be a potential blood biomarker for identifying advanced fibrosis in MASLD. Patients with the risk genotype PNPLA3 I148M G/G had higher levels of activin A proposing activin A as a contributor of the transition from simple steatosis to a fibrotic phenotype.
Collapse
Affiliation(s)
- Cecilia Jönsson
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Martin Bergram
- Division of Prevention, Rehabilitation and Community Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Stergios Kechagias
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Patrik Nasr
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Division of Diagnostics and Specialist Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
39
|
Vacca M, Kamzolas I, Harder LM, Oakley F, Trautwein C, Hatting M, Ross T, Bernardo B, Oldenburger A, Hjuler ST, Ksiazek I, Lindén D, Schuppan D, Rodriguez-Cuenca S, Tonini MM, Castañeda TR, Kannt A, Rodrigues CMP, Cockell S, Govaere O, Daly AK, Allison M, Honnens de Lichtenberg K, Kim YO, Lindblom A, Oldham S, Andréasson AC, Schlerman F, Marioneaux J, Sanyal A, Afonso MB, Younes R, Amano Y, Friedman SL, Wang S, Bhattacharya D, Simon E, Paradis V, Burt A, Grypari IM, Davies S, Driessen A, Yashiro H, Pors S, Worm Andersen M, Feigh M, Yunis C, Bedossa P, Stewart M, Cater HL, Wells S, Schattenberg JM, Anstee QM, Tiniakos D, Perfield JW, Petsalaki E, Davidsen P, Vidal-Puig A. An unbiased ranking of murine dietary models based on their proximity to human metabolic dysfunction-associated steatotic liver disease (MASLD). Nat Metab 2024; 6:1178-1196. [PMID: 38867022 PMCID: PMC11199145 DOI: 10.1038/s42255-024-01043-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/08/2024] [Indexed: 06/14/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease, encompasses steatosis and metabolic dysfunction-associated steatohepatitis (MASH), leading to cirrhosis and hepatocellular carcinoma. Preclinical MASLD research is mainly performed in rodents; however, the model that best recapitulates human disease is yet to be defined. We conducted a wide-ranging retrospective review (metabolic phenotype, liver histopathology, transcriptome benchmarked against humans) of murine models (mostly male) and ranked them using an unbiased MASLD 'human proximity score' to define their metabolic relevance and ability to induce MASH-fibrosis. Here, we show that Western diets align closely with human MASH; high cholesterol content, extended study duration and/or genetic manipulation of disease-promoting pathways are required to intensify liver damage and accelerate significant (F2+) fibrosis development. Choline-deficient models rapidly induce MASH-fibrosis while showing relatively poor translatability. Our ranking of commonly used MASLD models, based on their proximity to human MASLD, helps with the selection of appropriate in vivo models to accelerate preclinical research.
Collapse
Affiliation(s)
- Michele Vacca
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy.
- Laboratory of Liver Metabolism and MASLD, Roger Williams Institute of Hepatology, London, UK.
| | - Ioannis Kamzolas
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lea Mørch Harder
- Research and Early Development, Novo Nordisk A/S, Måløv, Copenhagen, Denmark
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Maximilian Hatting
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Trenton Ross
- Internal Medicine research Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Barbara Bernardo
- Internal Medicine research Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Anouk Oldenburger
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | | | - Iwona Ksiazek
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Daniel Lindén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
| | | | - Maria Manuela Tonini
- Luxembourg Institute of Health, Translational Medicine Operations Hub, Dudelange, Luxembourg
| | - Tamara R Castañeda
- R&D Diabetes & Portfolio Innovation and Excellence, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, Frankfurt, Germany
| | - Aimo Kannt
- R&D Diabetes, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Fraunhofer Innovation Center TheraNova and Goethe University, Frankfurt, Germany
| | - Cecília M P Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Simon Cockell
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Olivier Govaere
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ann K Daly
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Michael Allison
- Liver Unit, Cambridge University Hospitals NHS Foundation Trust & Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | | | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immunotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anna Lindblom
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Stephanie Oldham
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gaithersburg, MD, USA
| | - Anne-Christine Andréasson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Franklin Schlerman
- Inflammation and Immunology Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | | | - Arun Sanyal
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Marta B Afonso
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ramy Younes
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Yuichiro Amano
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric Simon
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Valérie Paradis
- Department of Imaging and Pathology, Université Paris Diderot and Hôpital Beaujon, Paris, France
| | - Alastair Burt
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Ioanna Maria Grypari
- Department of Pathology, Aretaeion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Susan Davies
- Department of Cellular Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ann Driessen
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
- Department of Molecular Imaging, Pathology, Radiotherapy, Oncology. Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Hiroaki Yashiro
- Research, Takeda Pharmaceuticals Company Limited, Cambridge, MA, USA
| | | | | | | | - Carla Yunis
- Pfizer, Inc.; Internal Medicine and Hospital, Pfizer Research and Development, Lake Mary, FL, USA
| | - Pierre Bedossa
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- LiverPat, Paris, France
| | | | | | - Sara Wells
- Mary Lyon Centre, MRC Harwell, Harwell Campus, Oxford, UK
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Medical Centre, Homburg, Germany
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
- Department of Pathology, Aretaeion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - James W Perfield
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK.
| | - Peter Davidsen
- Research and Early Development, Novo Nordisk A/S, Måløv, Copenhagen, Denmark.
- Ferring Pharmaceuticals A/S, International PharmaScience Center, Copenhagen, Denmark.
| | - Antonio Vidal-Puig
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigacion Principe Felipe, Valencia, Spain.
| |
Collapse
|
40
|
Tsutsumi T, Kawaguchi T, Fujii H, Kamada Y, Takahashi H, Kawanaka M, Sumida Y, Iwaki M, Hayashi H, Toyoda H, Oeda S, Hyogo H, Morishita A, Munekage K, Kawata K, Sawada K, Maeshiro T, Tobita H, Yoshida Y, Naito M, Araki A, Arakaki S, Noritake H, Ono M, Masaki T, Yasuda S, Tomita E, Yoneda M, Tokushige A, Ueda S, Aishima S, Nakajima A, Okanoue T. Hepatic inflammation and fibrosis are profiles related to mid-term mortality in biopsy-proven MASLD: A multicenter study in Japan. Aliment Pharmacol Ther 2024; 59:1559-1570. [PMID: 38651312 DOI: 10.1111/apt.17995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/20/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024]
Abstract
AIMS A multi-stakeholder consensus has proposed MASLD (metabolic dysfunction-associated steatotic liver disease). We aimed to investigate the pathological findings related to the mid-term mortality of patients with biopsy-proven MASLD in Japan. METHODS We enrolled 1349 patients with biopsy-proven MASLD. The observational period was 8010 person years. We evaluated independent factors associated with mortality in patients with MASLD by Cox regression analysis. We also investigated pathological profiles related to mortality in patients with MASLD using data-mining analysis. RESULTS The prevalence of MASH and stage 3/4 fibrosis was observed in 65.6% and 17.4%, respectively. Forty-five patients with MASLD died. Of these, liver-related events were the most common cause at 40% (n = 18), followed by extrahepatic malignancies at 26.7% (n = 12). Grade 2/3 lobular inflammation and stage 3/4 fibrosis had a 1.9-fold and 1.8-fold risk of mortality, respectively. In the decision-tree analysis, the profiles with the worst prognosis were characterised by Grade 2/3 hepatic inflammation, along with advanced ballooning (grade 1/2) and fibrosis (stage 3/4). This profile showed a mortality at 8.3%. Furthermore, the random forest analysis identified that hepatic fibrosis and inflammation were the first and second responsible factors for the mid-term prognosis of patients with MASLD. CONCLUSIONS In patients with biopsy-proven MASLD, the prevalence of MASH and advanced fibrosis was approximately 65% and 20%, respectively. The leading cause of mortality was liver-related events. Hepatic inflammation and fibrosis were significant factors influencing mid-term mortality. These findings highlight the importance of targeting inflammation and fibrosis in the management of patients with MASLD.
Collapse
Affiliation(s)
- Tsubasa Tsutsumi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hideki Fujii
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Miwa Kawanaka
- Department of General Internal Medicine2, Kawasaki Medical Center, Okayama, Japan
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare, Minato-ku, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideki Hayashi
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Satoshi Oeda
- Liver Center, Saga Medical School, Saga University, Saga, Japan
- Department of Laboratory Medicine, Saga University Hospital, Saga, Japan
| | | | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kensuke Munekage
- Department of Gastroenterology, Hata Kenmin Hospital, Sukumo, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Tatsuji Maeshiro
- First Department of Internal Medicine, Division of Infectious, Respiratory, and Digestive Medicine, University of the Ryukyus Graduate School of Medicine, Nishihara, Japan
| | - Hiroshi Tobita
- Department of Pathology, Shimane University Hospital, Izumo, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Osaka, Japan
| | - Masafumi Naito
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Osaka, Japan
| | - Asuka Araki
- Department of Pathology, Shimane University Hospital, Izumo, Japan
| | - Shingo Arakaki
- First Department of Internal Medicine, Division of Infectious, Respiratory, and Digestive Medicine, University of the Ryukyus Graduate School of Medicine, Nishihara, Japan
| | - Hidenao Noritake
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Masafumi Ono
- Division of Innovative Medicine for Hepatobiliary & Pancreatology, Faculty of Medicine, Kagawa University, Kita, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Satoshi Yasuda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Eiichi Tomita
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akihiro Tokushige
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Shinichi Aishima
- Department of Scientific Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | |
Collapse
|
41
|
Fan M, Song E, Zhang Y, Zhang P, Huang B, Yan K, Yang W, Chakrabarti S, Mahajan H, Yan S, Xu Y, Hua S, Liu W, Wang C, Xu A, Ye D. Metabolic Dysfunction-Associated Steatohepatitis Detected by Neutrophilic Crown-Like Structures in Morbidly Obese Patients: A Multicenter and Clinicopathological Study. RESEARCH (WASHINGTON, D.C.) 2024; 7:0382. [PMID: 38812532 PMCID: PMC11134285 DOI: 10.34133/research.0382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is the progressive form of metabolic dysfunction-associated steatotic liver disease (MASLD), and closely associated with a high risk of liver-related morbidity and mortality. Although enhanced neutrophil infiltration of the liver is a histological hallmark of MASH, the morphological pattern of hepatic neutrophils and their relevance to the definition of MASH remain unknown. This clinicopathological study aimed to determine the association of neutrophilic crown-like structures (CLSs) in liver biopsies and evaluate their relevance to the histological diagnosis of MASH. A total of 483 morbidly obese adults who underwent bariatric surgery were recruited. Neutrophilic CLSs in liver biopsies were detected by immunohistochemistry for neutrophil elastase and proteinase 3. All participants were classified into 4 histological subgroups: no MASLD (118, 24.4%), MASLD (76, 15.7%), borderline MASH (185, 38.3%), and definite MASH (104, 21.5%). In the discovery cohort (n = 379), the frequency of neutrophilic CLSs increased in line with the severity of liver disease. The number of neutrophilic CLSs was positively correlated with established histological characteristics of MASH. At a cutoff value of <0.3 per 20× microscopic field, the number of neutrophilic CLSs yielded a robust diagnostic accuracy to discriminate no MASLD and MASLD from borderline MASH and definite MASH; a cutoff at >1.3 per 20× microscopic field exhibited a statistically significant accuracy to distinguish definite MASH from other groups (no MASLD, MASLD, and borderline MASH). The significance of neutrophilic CLSs in identifying borderline MASH and definite MASH was confirmed in an external validation cohort (n = 104). The frequency of neutrophilic CLSs was significantly higher than that of macrophagic CLSs. In conclusion, neutrophilic CLSs in the liver represent a typical histological characteristic of MASH and may serve as a promising indicator to improve the diagnostic accuracy of MASH during histological assessment of liver biopsies.
Collapse
Affiliation(s)
- Mengqi Fan
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Erfei Song
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yuying Zhang
- Department of Obstetrics, Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Pengfei Zhang
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bing Huang
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kaixuan Yan
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wah Yang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Hema Mahajan
- Institute of Clinical Pathology and Medical Research, Pathology West, NSW Health Pathology, Sydney, NSW 2145, Australia
| | - Sen Yan
- Dr. Everett Chalmers Hospital, Fredericton, NB, Canada
| | - Ying Xu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuang Hua
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wei Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cunchuan Wang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dewei Ye
- Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
42
|
Taru MG, Tefas C, Neamti L, Minciuna I, Taru V, Maniu A, Rusu I, Petrushev B, Procopciuc LM, Leucuta DC, Procopet B, Ferri S, Lupsor-Platon M, Stefanescu H. FAST and Agile-the MASLD drift: Validation of Agile 3+, Agile 4 and FAST scores in 246 biopsy-proven NAFLD patients meeting MASLD criteria of prevalent caucasian origin. PLoS One 2024; 19:e0303971. [PMID: 38781158 PMCID: PMC11115280 DOI: 10.1371/journal.pone.0303971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND MASLD is a prevalent chronic liver condition with substantial clinical implications. This study aimed to assess the effectiveness of three new, elastography-based, scoring systems for advanced fibrosis ≥F3 (Agile 3+), cirrhosis F4 (Agile 4), and fibrotic NASH: NASH + NAS ≥4 + F≥2 (FAST score), in a cohort of biopsy-proven NAFLD meeting MASLD criteria. Our secondary aim was to compare their diagnostic performances with those of other fibrosis prediction tools: LSM-VCTE alone, and common, easily available scores (FIB-4 or APRI). METHODS Single-center, retrospective study, on consecutive patients with baseline laboratory tests, liver biopsy, and reliable LSM-VCTE measurements. The discrimination between tests was evaluated by analyzing the AUROCs. Dual cut-off approaches were applied to rule-out and rule-in ≥F3, F4 and fibrotic NASH. We tested previously reported cut-off values and provided our best thresholds to achieve Se ≥85%, Se ≥90%, and Sp ≥90%, Sp ≥95%. RESULTS Among 246 patients, 113 (45.9%) were women, and 75 (30.5%) presented diabetes. Agile 3+ and Agile 4 demonstrated excellent performance in identifying ≥F3 and F4, achieving AUROCs of 0.909 and 0.968, while the FAST score yielded acceptable results in distinguishing fibrotic NASH. When compared to FIB-4 and LSM-VCTE, both Agile 3+ and Agile 4 performed better than FIB-4 and had a similar performance to LSM-VCTE, but with higher diagnostic accuracy, hence reducing the grey zone. CONCLUSION Agile 3+ and Agile 4 are reliable, non-invasive tests for identifying advanced fibrosis or cirrhosis in MASLD patients, while FAST score demonstrates moderate performance in identifying fibrotic NASH.
Collapse
Affiliation(s)
- Madalina-Gabriela Taru
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Cristian Tefas
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Lidia Neamti
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Iulia Minciuna
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Vlad Taru
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Anca Maniu
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Ioana Rusu
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Bobe Petrushev
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Lucia Maria Procopciuc
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
| | - Dan Corneliu Leucuta
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
| | - Bogdan Procopet
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Silvia Ferri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
| | - Monica Lupsor-Platon
- Faculty of Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| | - Horia Stefanescu
- Regional Institute of Gastroenterology and Hepatology "Octavian Fodor", Cluj-Napoca, Cluj, Romania
| |
Collapse
|
43
|
Cooreman MP, Butler J, Giugliano RP, Zannad F, Dzen L, Huot-Marchand P, Baudin M, Beard DR, Junien JL, Broqua P, Abdelmalek MF, Francque SM. The pan-PPAR agonist lanifibranor improves cardiometabolic health in patients with metabolic dysfunction-associated steatohepatitis. Nat Commun 2024; 15:3962. [PMID: 38730247 PMCID: PMC11087475 DOI: 10.1038/s41467-024-47919-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
Lanifibranor, a pan-PPAR agonist, improves liver histology in patients with metabolic dysfunction-associated steatohepatitis (MASH), who have poor cardiometabolic health (CMH) and cardiovascular events as major mortality cause. NATIVE trial secondary and exploratory outcomes (ClinicalTrials.gov NCT03008070) were analyzed for the effect of lanifibranor on IR, lipid and glucose metabolism, systemic inflammation, blood pressure (BP), hepatic steatosis (imaging and histological grading) for all patients of the original analysis. With lanifibranor, triglycerides, HDL-C, apolipoproteins, insulin, HOMA-IR, HbA1c, fasting glucose (FG), hs-CRP, ferritin, diastolic BP and steatosis improved significantly, independent of diabetes status: most patients with prediabetes returned to normal FG levels. Significant adiponectin increases correlated with hepatic and CMH marker improvement; patients had an average weight gain of 2.5 kg, with 49% gaining ≥2.5% weight. Therapeutic benefits were similar regardless of weight change. Here, we show that effects of lanifibranor on liver histology in MASH are accompanied with CMH improvement, indicative of potential cardiovascular clinical benefits.
Collapse
Affiliation(s)
- Michael P Cooreman
- Research and Development, Inventiva, New York, NY, USA.
- Research and Development, Inventiva, Daix, France.
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
| | - Robert P Giugliano
- Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Faiez Zannad
- Centre d'Investigations Cliniques Plurithématique 1433, Université de Lorraine, Nancy, France
| | - Lucile Dzen
- Research and Development, Inventiva, New York, NY, USA
- Research and Development, Inventiva, Daix, France
| | - Philippe Huot-Marchand
- Research and Development, Inventiva, New York, NY, USA
- Research and Development, Inventiva, Daix, France
| | - Martine Baudin
- Research and Development, Inventiva, New York, NY, USA
- Research and Development, Inventiva, Daix, France
| | | | - Jean-Louis Junien
- Research and Development, Inventiva, New York, NY, USA
- Research and Development, Inventiva, Daix, France
| | - Pierre Broqua
- Research and Development, Inventiva, New York, NY, USA
- Research and Development, Inventiva, Daix, France
| | - Manal F Abdelmalek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Sven M Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| |
Collapse
|
44
|
Dioguardi Burgio M, Castera L, Oufighou M, Rautou PE, Paradis V, Bedossa P, Sartoris R, Ronot M, Bodard S, Garteiser P, Van Beers B, Valla D, Vilgrain V, Correas JM. Prospective Comparison of Attenuation Imaging and Controlled Attenuation Parameter for Liver Steatosis Diagnosis in Patients With Nonalcoholic Fatty Liver Disease and Type 2 Diabetes. Clin Gastroenterol Hepatol 2024; 22:1005-1013.e27. [PMID: 38072287 DOI: 10.1016/j.cgh.2023.11.034] [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: 06/20/2023] [Revised: 10/31/2023] [Accepted: 11/26/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND & AIMS Similarly to the controlled attenuation parameter (CAP), the ultrasound-based attenuation imaging (ATI) can quantify hepatic steatosis. We prospectively compared the performance of ATI and CAP for the diagnosis of hepatic steatosis in patients with type 2 diabetes and nonalcoholic fatty liver disease using histology and magnetic resonance imaging-proton density fat fraction (MRI-PDFF) as references. METHODS Patients underwent ATI and CAP measurement, MRI, and biopsy on the same day. Steatosis was classified as S0, S1, S2, and S3 on histology (<5%, 5%-33%, 33%-66%, and >66%, respectively) while the thresholds of 6.4%, 17.4%, and 22.1%, respectively, were used for MRI-PDFF. The area under the curve (AUC) of ATI and CAP was compared using a DeLong test. RESULTS Steatosis could be evaluated in 191 and 187 patients with MRI-PDFF and liver biopsy, respectively. For MRI-PDFF steatosis, the AUC of ATI and CAP were 0.86 (95% confidence interval [CI], 0.81-0.91) vs 0.69 (95% CI, 0.62-0.75) for S0 vs S1-S3 (P = .02) and 0.71 (95% CI, 0.64-0.77) vs 0.69 (95% CI, 0.61-0.75) for S0-S1 vs S2-S3 (P = .60), respectively. For histological steatosis, the AUC of ATI and CAP were 0.92 (95% CI, 0.87-0.95) vs 0.95 (95% CI, 0.91-0.98) for S0 vs S1-S3 (P = .64) and 0.79 (95% CI, 0.72-0.84) vs 0.76 (95% CI, 0.69-0.82) for S0-S1 vs S2-S3 (P = .61), respectively. CONCLUSION ATI may be used as an alternative to CAP for the diagnosis and quantification of steatosis, in patients with type 2 diabetes and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Marco Dioguardi Burgio
- Department of Radiology, Hôpital Beaujon, AP-HP Nord, Clichy, France; Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France.
| | - Laurent Castera
- Departement of Hepatology, Hospital Beaujon, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Mehdi Oufighou
- Department of Radiology, Hôpital Beaujon, AP-HP Nord, Clichy, France
| | - Pierre-Emmanuel Rautou
- Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France; Service d'Hépatologie, AP-HP, Hôpital Beaujon, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Clichy, France
| | - Valérie Paradis
- Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France; Department of Pathology, Hôpital Beaujon, AP-HP Nord, Clichy, France
| | - Pierre Bedossa
- Department of Pathology, Hôpital Beaujon, AP-HP Nord, Clichy, France
| | - Riccardo Sartoris
- Department of Radiology, Hôpital Beaujon, AP-HP Nord, Clichy, France
| | - Maxime Ronot
- Department of Radiology, Hôpital Beaujon, AP-HP Nord, Clichy, France; Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France
| | - Sylvain Bodard
- Department of Adult Radiology, Necker University Hospital, AP-HP, Paris, France; Université Paris Cité, Paris, France
| | - Philippe Garteiser
- Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France
| | - Bernard Van Beers
- Department of Radiology, Hôpital Beaujon, AP-HP Nord, Clichy, France; Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France
| | - Dominique Valla
- Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France; Service d'Hépatologie, AP-HP, Hôpital Beaujon, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Clichy, France
| | - Valérie Vilgrain
- Department of Radiology, Hôpital Beaujon, AP-HP Nord, Clichy, France; Université Paris Cité, INSERM, Centre de Recherche sur L'inflammation, Paris, France
| | - Jean Michel Correas
- Department of Adult Radiology, Necker University Hospital, AP-HP, Paris, France; Université Paris Cité, Paris, France; Sorbonne Université, CNRS, INSERM Laboratoire d'Imagerie Biomédicale, Paris, France
| |
Collapse
|
45
|
Zhao X, Ma R, Abulikemu A, Qi Y, Liu X, Wang J, Xu K, Guo C, Li Y. Proteomics revealed composition- and size-related regulators for hepatic impairments induced by silica nanoparticles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:170584. [PMID: 38309355 DOI: 10.1016/j.scitotenv.2024.170584] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Along with the growing production and application of silica nanoparticles (SiNPs), increased human exposure and ensuing safety evaluation have progressively attracted concern. Accumulative data evidenced the hepatic injuries upon SiNPs inhalation. Still, the understanding of the hepatic outcomes resulting from SiNPs exposure, and underlying mechanisms are incompletely elucidated. Here, SiNPs of two sizes (60 nm and 300 nm) were applied to investigate their composition- and size-related impacts on livers of ApoE-/- mice via intratracheal instillation. Histopathological and biochemical analysis indicated SiNPs promoted inflammation, lipid deposition and fibrosis in the hepatic tissue, accompanied by increased ALT, AST, TC and TG. Oxidative stress was activated upon SiNPs stimuli, as evidenced by the increased hepatic ROS, MDA and declined GSH/GSSG. Of note, these alterations were more dramatic in SiNPs with a smaller size (SiNPs-60) but the same dosage. LC-MS/MS-based quantitative proteomics unveiled changes in mice liver protein profiles, and filtered out particle composition- or size-related molecules. Interestingly, altered lipid metabolism and oxidative damage served as two critical biological processes. In accordance with correlation analysis and liver disease-targeting prediction, a final of 10 differentially expressed proteins (DEPs) were selected as key potential targets attributable to composition- (4 molecules) and size-related (6 molecules) liver impairments upon SiNPs stimuli. Overall, our study provided strong laboratory evidence for a comprehensive understanding of the harmful biological effects of SiNPs, which was crucial for toxicological evaluation to ensure nanosafety.
Collapse
Affiliation(s)
- Xinying Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Ru Ma
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Alimire Abulikemu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yi Qi
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xiaoying Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Ji Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Kun Xu
- School of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Caixia Guo
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
46
|
Savari F, Mard SA. Nonalcoholic steatohepatitis: A comprehensive updated review of risk factors, symptoms, and treatment. Heliyon 2024; 10:e28468. [PMID: 38689985 PMCID: PMC11059522 DOI: 10.1016/j.heliyon.2024.e28468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 05/02/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a subtype of nonalcoholic fatty liver disease and a progressive and chronic liver disorder with a significant risk for the development of liver-related morbidity and mortality. The complex and multifaceted pathophysiology of NASH makes its management challenging. Early identification of symptoms and management of patients through lifestyle modification is essential to prevent the development of advanced liver disease. Despite the increasing prevalence of NASH, there is no FDA-approved treatment for this disease. Currently, medications targeting metabolic disease risk factors and some antifibrotic medications are used for NASH patients but are not sufficiently effective. The beneficial effects of different drugs and phytochemicals represent new avenues for the development of safer and more effective treatments for NASH. In this review, different risk factors, clinical symptoms, diagnostic methods of NASH, and current treatment strategies for the management of patients with NASH are reviewed.
Collapse
Affiliation(s)
- Feryal Savari
- Department of Medical Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Seyed Ali Mard
- Clinical Sciences Research Institute, Alimentary Tract Research Center, Department of Physiology, The School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
47
|
Iwaki M, Fujii H, Hayashi H, Toyoda H, Oeda S, Hyogo H, Kawanaka M, Morishita A, Munekage K, Kawata K, Tsutsumi T, Sawada K, Maeshiro T, Tobita H, Yoshida Y, Naito M, Araki A, Arakaki S, Kawaguchi T, Noritake H, Ono M, Masaki T, Yasuda S, Tomita E, Yoneda M, Tokushige A, Kamada Y, Takahashi H, Ueda S, Aishima S, Sumida Y, Nakajima A, Okanoue T, Japan Study Group of Nonalcoholic Fatty Liver Disease (JSG-NAFLD). Prognosis of biopsy-confirmed metabolic dysfunction- associated steatotic liver disease: A sub-analysis of the CLIONE study. Clin Mol Hepatol 2024; 30:225-234. [PMID: 38263684 PMCID: PMC11016478 DOI: 10.3350/cmh.2023.0515] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND/AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) was recently proposed as an alternative disease concept to nonalcoholic fatty liver disease (NAFLD). We aimed to investigate the prognosis of patients with biopsy-confirmed MASLD using data from a multicenter study. METHODS This was a sub-analysis of the Clinical Outcome Nonalcoholic Fatty Liver Disease (CLIONE) study that included 1,398 patients with NAFLD. Liver biopsy specimens were pathologically diagnosed and histologically scored using the NASH Clinical Research Network system, the FLIP algorithm, and the SAF score. Patients who met at least one cardiometabolic criterion were diagnosed with MASLD. RESULTS Approximately 99% of cases (n=1,381) were classified as MASLD. Patients with no cardiometabolic risk (n=17) had a significantly lower BMI than patients with MASLD (20.9 kg/m2 vs. 28.0 kg/m2, P<0.001), in addition to significantly lower levels of inflammation, ballooning, NAFLD activity score, and fibrosis stage based on liver histology. These 17 patients had a median follow-up of 5.9 years, equivalent to 115 person-years, with no deaths, liver-related events, cardiovascular events, or extrahepatic cancers. The results showed that the prognosis for pure MASLD was similar to that for the original CLIONE cohort, with 47 deaths and one patient who underwent orthotopic liver transplantation. The leading cause of death was extrahepatic cancer (n=10), while the leading causes of liver-related death were liver failure (n=9), hepatocellular carcinoma (n=8), and cholangiocarcinoma (n=4). CONCLUSION Approximately 99% of NAFLD cases were considered MASLD based on the 2023 liver disease nomenclature. The NAFLD-only group, which is not encompassed by MASLD, had a relatively mild histopathologic severity and a favorable prognosis. Consequently, the prognosis of MASLD is similar to that previously reported for NAFLD.
Collapse
Affiliation(s)
- Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideki Fujii
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hideki Hayashi
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Satoshi Oeda
- Liver Center and Department of Laboratory Medicine, Saga University Hospital, Saga, Japan
| | | | - Miwa Kawanaka
- Department of General Internal Medicine2, Kawasaki Medical Center, Kawasaki Medical School, Okayama, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kensuke Munekage
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
- Department of Gastroenterology, Kochi Prefectural Hata Kenmin Hospital, Kochi, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Tsubasa Tsutsumi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Tatsuji Maeshiro
- Department of Gastroenterology, Urasoe General Hospital, Okinawa, Japan
| | - Hiroshi Tobita
- Department of Hepatology, Shimane University Hospital, Shimane, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Osaka, Japan
| | - Masafumi Naito
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Osaka, Japan
| | - Asuka Araki
- Division of Pathology, Shimane University Hospital, Shimane, Japan
| | - Shingo Arakaki
- Department of Gastroenterology, Urasoe General Hospital, Okinawa, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hidenao Noritake
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Masafumi Ono
- Division of Innovative Medicine for Hepatobiliary & Pancreatology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Satoshi Yasuda
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Eiichi Tomita
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Akihiro Tokushige
- Department of Clinical Pharmacology and Therapeutics School of Medicine University of the Ryukyus, Okinawa, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University, Graduate School of Medicine, Osaka, Japan
| | | | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics School of Medicine University of the Ryukyus, Okinawa, Japan
| | - Shinichi Aishima
- Department of Scientific Pathology Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare, Tokyo, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Japan Study Group of Nonalcoholic Fatty Liver Disease (JSG-NAFLD)
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, Gifu, Japan
- Department of Gastroenterology, Ogaki Municipal Hospital, Ogaki, Japan
- Liver Center and Department of Laboratory Medicine, Saga University Hospital, Saga, Japan
- Hyogo Life Care Clinic Hiroshima, Hiroshima, Japan
- Department of General Internal Medicine2, Kawasaki Medical Center, Kawasaki Medical School, Okayama, Japan
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
- Department of Gastroenterology, Kochi Prefectural Hata Kenmin Hospital, Kochi, Japan
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Shizuoka, Japan
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
- Department of Gastroenterology, Urasoe General Hospital, Okinawa, Japan
- Department of Hepatology, Shimane University Hospital, Shimane, Japan
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Osaka, Japan
- Division of Pathology, Shimane University Hospital, Shimane, Japan
- Division of Innovative Medicine for Hepatobiliary & Pancreatology, Faculty of Medicine, Kagawa University, Kagawa, Japan
- Department of Clinical Pharmacology and Therapeutics School of Medicine University of the Ryukyus, Okinawa, Japan
- Department of Advanced Metabolic Hepatology, Osaka University, Graduate School of Medicine, Osaka, Japan
- Liver Center, Saga University Hospital, Saga, Japan
- Department of Scientific Pathology Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Graduate School of Healthcare Management, International University of Healthcare and Welfare, Tokyo, Japan
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| |
Collapse
|
48
|
Herrera-Marcos LV, Martínez-Beamonte R, Arnal C, Barranquero C, Puente-Lanzarote JJ, Lou-Bonafonte JM, Gonzalo-Romeo G, Mocciaro G, Jenkins B, Surra JC, Rodríguez-Yoldi MJ, Alastrué-Vera V, Letosa J, García-Gil A, Güemes A, Koulman A, Osada J. Lipidomic signatures discriminate subtle hepatic changes in the progression of porcine nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2024; 326:G411-G425. [PMID: 38375587 DOI: 10.1152/ajpgi.00264.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/21/2024]
Abstract
Recently, the development of nonalcoholic steatohepatitis (NASH) in common strains of pigs has been achieved using a diet high in saturated fat, fructose, cholesterol, and cholate and deficient in choline and methionine. The aim of the present work was to characterize the hepatic and plasma lipidomic changes that accompany the progression of NASH and its reversal by switching pigs back to a chow diet. One month of this extreme steatotic diet was sufficient to induce porcine NASH. The lipidomic platform using liquid chromatography-mass spectrometry analyzed 467 lipid species. Seven hepatic phospholipids [PC(30:0), PC(32:0), PC(33:0), PC(33:1), PC(34:0), PC(34:3) and PC(36:2)] significantly discriminated the time of dietary exposure, and PC(30:0), PC(33:0), PC(33:1) and PC(34:0) showed rapid adaptation in the reversion period. Three transcripts (CS, MAT1A, and SPP1) showed significant changes associated with hepatic triglycerides and PC(33:0). Plasma lipidomics revealed that these species [FA 16:0, FA 18:0, LPC(17:1), PA(40:5), PC(37:1), TG(45:0), TG(47:2) and TG(51:0)] were able to discriminate the time of dietary exposure. Among them, FA 16:0, FA 18:0, LPC(17:1) and PA(40:5) changed the trend in the reversion phase. Plasma LDL-cholesterol and IL12P40 were good parameters to study the progression of NASH, but their capacity was surpassed by hepatic [PC(33:0), PC(33:1), and PC(34:0)] or plasma lipid [FA 16:0, FA 18:0, and LPC(17:1)] species. Taken together, these lipid species can be used as biomarkers of metabolic changes in the progression and regression of NASH in this model. The lipid changes suggest that the development of NASH also affects peripheral lipid metabolism.NEW & NOTEWORTHY A NASH stage was obtained in crossbred pigs. Hepatic [PC(33:0), PC(33:1) and PC(34:0)] or plasma [FA 16:0, FA 18:0 and LPC(17:1)] species were sensitive parameters to detect subtle changes in development and regression of nonalcoholic steatohepatitis (NASH). These findings may delineate the liquid biopsy to detect subtle changes in progression or in treatments. Furthermore, phospholipid changes according to the insult-inducing NASH may play an important role in accepting or rejecting fatty livers in transplantation.
Collapse
Affiliation(s)
- Luis V Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan J Puente-Lanzarote
- Servicio de Bioquímica Clínica, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - José M Lou-Bonafonte
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo Gonzalo-Romeo
- Servicio General de Apoyo a la Investigación, División de Experimentación Animal, Universidad de Zaragoza, Zaragoza, Spain
| | - Gabriele Mocciaro
- Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Benjamin Jenkins
- Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Huesca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jesús Letosa
- Industrial Zootécnica Aragonesa S.L. (INZAR, S.L.), Zaragoza, Spain
| | - Agustín García-Gil
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Antonio Güemes
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
49
|
Suppli MP, Høgedal A, Bagger JI, Chabanova E, van Hall G, Forman JL, Christensen MB, Albrechtsen NJW, Holst JJ, Knop FK. Signs of Glucagon Resistance After a 2-Week Hypercaloric Diet Intervention. J Clin Endocrinol Metab 2024; 109:955-967. [PMID: 37967235 DOI: 10.1210/clinem/dgad666] [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: 09/21/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023]
Abstract
CONTEXT Hyperglucagonemia is observed in individuals with obesity and contributes to the hyperglycemia of patients with type 2 diabetes. Hyperglucagonemia may develop due to steatosis-induced hepatic glucagon resistance resulting in impaired hepatic amino acid turnover and ensuing elevations of circulating glucagonotropic amino acids. OBJECTIVE We evaluated whether glucagon resistance could be induced in healthy individuals by a hypercaloric diet intervention designed to increase hepatic fat content. METHODS We recruited 20 healthy male individuals to follow a hypercaloric diet and a sedentary lifestyle for 2 weeks. Amino acid concentrations in response to infusion of glucagon were assessed during a pancreatic clamp with somatostatin and basal insulin. The reversibility of any metabolic changes was assessed 8 weeks after the intervention. Hepatic steatosis was assessed by magnetic resonance spectroscopy. RESULTS The intervention led to increased hepatic fat content (382% [206%; 705%], P < .01). Glucagon infusion led to a decrease in the concentration of total amino acids on all experimental days, but the percentage change in total amino acids was reduced (-2.5% ± 0.5% vs -0.2% ± 0.7%, P = .015) and the average slope of the decline in the total amino acid concentration was less steep (-2.0 ± 1.2 vs -1.2 ± 0.3 μM/min, P = .016) after the intervention compared to baseline. The changes were normalized at follow-up. CONCLUSION Our results indicate that short-term unhealthy behavior, which increases hepatic fat content, causes a reversible resistance to the effect of glucagon on amino acid concentrations in healthy individuals, which may explain the hyperglucagonemia associated with obesity and diabetes.
Collapse
Affiliation(s)
- Malte Palm Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Astrid Høgedal
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Jonatan Ising Bagger
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Steno Diabetes Center Copenhagen, DK-2730 Herlev, Denmark
| | - Elizaveta Chabanova
- Department of Radiology, Herlev Hospital, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Gerrit van Hall
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Julie Lyng Forman
- Section of Biostatistics, Department of Public Health, University of Copenhagen, DK-1353 Copenhagen, Denmark
| | - Mikkel Bring Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
| | - Nicolai Jacob Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Filip Krag Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Steno Diabetes Center Copenhagen, DK-2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
50
|
Boesch M, Lindhorst A, Feio-Azevedo R, Brescia P, Silvestri A, Lannoo M, Deleus E, Jaekers J, Topal H, Topal B, Ostyn T, Wallays M, Smets L, Van Melkebeke L, Härtlova A, Roskams T, Bedossa P, Verbeek J, Govaere O, Francque S, Sifrim A, Voet T, Rescigno M, Gericke M, Korf H, van der Merwe S. Adipose tissue macrophage dysfunction is associated with a breach of vascular integrity in NASH. J Hepatol 2024; 80:397-408. [PMID: 37977244 DOI: 10.1016/j.jhep.2023.10.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/03/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND & AIMS In non-alcoholic fatty liver disease (NAFLD), monocytes infiltrate visceral adipose tissue promoting local and hepatic inflammation. However, it remains unclear what drives inflammation and how the immune landscape in adipose tissue differs across the NAFLD severity spectrum. We aimed to assess adipose tissue macrophage (ATM) heterogeneity in a NAFLD cohort. METHODS Visceral adipose tissue macrophages from lean and obese patients, stratified by NAFLD phenotypes, underwent single-cell RNA sequencing. Adipose tissue vascular integrity and breaching was assessed on a protein level via immunohistochemistry and immunofluorescence to determine targets of interest. RESULTS We discovered multiple ATM populations, including resident vasculature-associated macrophages (ResVAMs) and distinct metabolically active macrophages (MMacs). Using trajectory analysis, we show that ResVAMs and MMacs are replenished by a common transitional macrophage (TransMac) subtype and that, during NASH, MMacs are not effectively replenished by TransMac precursors. We postulate an accessory role for MMacs and ResVAMs in protecting the adipose tissue vascular barrier, since they both interact with endothelial cells and localize around the vasculature. However, across the NAFLD severity spectrum, alterations occur in these subsets that parallel an adipose tissue vasculature breach characterized by albumin extravasation into the perivascular tissue. CONCLUSIONS NAFLD-related macrophage dysfunction coincides with a loss of adipose tissue vascular integrity, providing a plausible mechanism by which tissue inflammation is perpetuated in adipose tissue and downstream in the liver. IMPACT AND IMPLICATIONS Our study describes for the first time the myeloid cell landscape in human visceral adipose tissue at single-cell level within a cohort of well-characterized patients with non-alcoholic fatty liver disease. We report unique non-alcoholic steatohepatitis-specific transcriptional changes within metabolically active macrophages (MMacs) and resident vasculature-associated macrophages (ResVAMs) and we demonstrate their spatial location surrounding the vasculature. These dysfunctional transcriptional macrophage states coincided with the loss of adipose tissue vascular integrity, providing a plausible mechanism by which tissue inflammation is perpetuated in adipose tissue and downstream in the liver. Our study provides a theoretical basis for new therapeutic strategies to be directed towards reinstating the endogenous metabolic, homeostatic and cytoprotective functions of ResVAMs and MMacs, including their role in protecting vascular integrity.
Collapse
Affiliation(s)
- Markus Boesch
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | | | - Rita Feio-Azevedo
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Paola Brescia
- IRCCS Humanitas Research Hospital, Manzoni 56, 20089 Rozzano, Milan, Italy
| | | | | | - Ellen Deleus
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Joris Jaekers
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Halit Topal
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Baki Topal
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Tessa Ostyn
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, 3000 Leuven, Belgium
| | - Marie Wallays
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Lena Smets
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Lukas Van Melkebeke
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Anetta Härtlova
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Tania Roskams
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, 3000 Leuven, Belgium
| | - Pierre Bedossa
- Department of Pathology, Physiology and Imaging, Beaujon Hospital Paris Diderot University, Paris, France
| | - Jef Verbeek
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Olivier Govaere
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, 3000 Leuven, Belgium
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium; Translational Research in Inflammation and Immunology (TWI2N), Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Alejandro Sifrim
- KU Leuven Institute for Single Cell Omics (LISCO), 3000 Leuven, Belgium; Laboratory of Multi-omic Integrative Bioinformatics, Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Thierry Voet
- KU Leuven Institute for Single Cell Omics (LISCO), 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20072 Pieve Emanuele, Milan, Italy
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Hannelie Korf
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium.
| | - Schalk van der Merwe
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium.
| |
Collapse
|