1
|
Piecha F, Jahn B, Köntopf J, Koop A, Ozga A, Al‐Jawazneh A, Harberts A, Riedel C, Buggisch P, Benten D, Hübener P, Adam G, Huber S, Lohse AW, Bannas P, Kluwe J. Recompensation of Liver Cirrhosis by TIPS Reduces Epithelial Cell Death Markers, Translating Into Improved Clinical Outcome. Liver Int 2025; 45:e16156. [PMID: 39533838 PMCID: PMC11897859 DOI: 10.1111/liv.16156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS Portal hypertension is the main pathophysiological driver of decompensation in patients with liver cirrhosis. Epithelial cell death markers, m30 and m65, correlate with hepatic injury and predict outcomes across various stages of liver disease. We aim (i) to evaluate whether portal hypertension itself contributes to liver outcome-relevant epithelial injury, and (ii) to analyse the capacity of m30/m65 to predict outcome in patients receiving a transjugular intrahepatic portosystemic shunt (TIPS) for refractory ascites. METHODS Sixty-six patients undergoing TIPS placement for refractory ascites and 20 patients with compensated cirrhosis as controls were prospectively enrolled in this monocentric cohort study. Epithelial cell death markers were analysed pre-TIPS, as well as 1-3 and 6-9 months post-TIPS. The capacity of baseline levels of m30/m65 in predicting six-month transplant-free survival rates was analysed by multivariable Cox proportional hazards regression. RESULTS Levels of m30 and m65 were higher in patients with decompensated cirrhosis (pre-TIPS) compared with compensated cirrhosis (controls). Following correction of portal hypertension by TIPS and recompensation, both markers decreased over time, reaching levels comparable to patients with compensated cirrhosis. On multivariable analysis, pre-TIPS baseline levels of m30 and m65 were not predictive for six-month survival. CONCLUSION Correction of portal hypertension via TIPS reduces levels of epithelial cell death markers, indicating that portal hypertension is a driver of outcome-relevant, hepatic cell death in patients with decompensated cirrhosis. Baseline m30/m65 values do not affect six-month survival rates, which suggests that TIPS placement overcomes the unfavourable spontaneous prognosis otherwise indicated by elevated baseline m30/65 levels.
Collapse
Affiliation(s)
- Felix Piecha
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center for Infection Research (DZIF), Partner Site Hamburg‐Lübeck‐Borstel‐RiemsHamburgGermany
| | | | - Johannes Köntopf
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Anja Koop
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ann‐Kathrin Ozga
- Center for Experimental Medicine, Institute of Medical Biometry and EpidemiologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Amirah Al‐Jawazneh
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
| | - Aenne Harberts
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christoph Riedel
- Department of Diagnostic and Interventional Radiology and Nuclear MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Peter Buggisch
- Ifi‐Institute for Interdisciplinary MedicineHamburgGermany
| | - Daniel Benten
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of GastroenterologyAsklepios Hospital HarburgHamburgGermany
| | - Peter Hübener
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Samuel Huber
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ansgar W. Lohse
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Peter Bannas
- Department of Diagnostic and Interventional Radiology and Nuclear MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Johannes Kluwe
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
2
|
Askeland A, Rasmussen RW, Gjela M, Frøkjær JB, Højlund K, Mellergaard M, Handberg A. Non-invasive liver fibrosis markers are increased in obese individuals with non-alcoholic fatty liver disease and the metabolic syndrome. Sci Rep 2025; 15:10652. [PMID: 40148373 PMCID: PMC11950363 DOI: 10.1038/s41598-025-85508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/03/2025] [Indexed: 03/29/2025] Open
Abstract
The need for early non-invasive diagnostic tools for chronic liver fibrosis is growing, particularly in individuals with obesity, non-alcoholic fatty liver disease (NAFLD), and the metabolic syndrome (MetS) since prevalence of these conditions is increasing. This case-control study compared non-invasive liver fibrosis markers in obesity with NAFLD and MetS (NAFLD-MetS, n = 33), in obese (n = 28) and lean (n = 27) control groups. We used MRI (T1 relaxation times (T1) and liver stiffness), circulating biomarkers (CK18, PIIINP, and TIMP1), and algorithms (FIB-4 index, Forns score, FNI, and MACK3 score) to assess their potential in predicting liver fibrosis risk. We found that T1 (892 ± 81 ms vs. 818 ± 64 ms, p < 0.001), FNI (15 ± 12% vs. 9 ± 7%, p = 0.018), CK18 (166 ± 110 U/L vs. 113 ± 41 U/L, p = 0.019), and MACK3 (0.18 ± 0.15 vs. 0.05 ± 0.04, p < 0.001) were higher in the NAFLD-MetS group compared with the obese control group. Moreover, correlations were found between CK18 and FNI (r = 0.69, p < 0.001), CK18 and T1 (r = 0.41, p < 0.001), FNI and T1 (r = 0.33, p = 0.006), MACK3 and FNI (r = 0.79, p < 0.001), and MACK3 and T1 (r = 0.50, p < 0.001). We show that liver fibrosis markers are increased in obese individuals with NAFLD and MetS without clinical signs of liver fibrosis. More studies are needed to validate the use of these non-invasive biomarkers for early identification of liver fibrosis risk.
Collapse
Affiliation(s)
- Anders Askeland
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Mimoza Gjela
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Brøndum Frøkjær
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
3
|
Ye J, Lai J, Luo L, Zhou T, Sun Y, Zhong B. Cytokeratin 18 fragment in liver inflammation and fibrosis: Systematic review and meta-analysis. Clin Chim Acta 2025; 569:120147. [PMID: 39832704 DOI: 10.1016/j.cca.2025.120147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND This meta-analysis aimed to summarize the diagnostic accuracy and cut-off values of cytokeratin (CK) 18 measurements, specifically M30 and M65, as candidate biomarkers for the pathological evaluation of biopsy specimens used to stage liver inflammation and fibrosis in patients with chronic liver diseases. METHODS Databases were searched for studies collected up to January 11th, 2025. Pooled sensitivity, specificity, area under the receiver-operating characteristic curves, and mean cut-off values were calculated using random-effects models regardless of heterogeneity. A meta-regression analysis and subgroup analysis were performed to explore heterogeneity. RESULTS Sixty-three studies comprising 9137 patients were included. The summarized AUROC curve of CK18 M30 for the diagnosis of significant liver inflammation, fibrosis ≥F1, ≥F2, ≥F3, and =F4 according to the METAVIR score system were 0.82, 0.75, 0.78, 0.78 and 0.76, with mean cut-off values of 264.3, 188.0, 276.9, 322.8 and 169.4 U/L. For M65, the summarized AUROC curve for detecting significant liver inflammation, fibrosis ≥F1, ≥F2, and =F4 were 0.79, 0.70, 0.76, 0.64 and 0.72, with mean cut-off values of 541.1, 417.6, 500.1, 424.6 and 674.0 U/L. The subgroup analyses implied that ethnicity may be the primary factor related to heterogeneity in CK18 M30 when applied to detect significant inflammation. Asian patients had values 79.7 U/L higher than those of non-Asian patients (p = 0.0157). CONCLUSIONS CK18 M30 and M65 have clinically meaningful accuracy as alternative diagnostic tools for determining liver inflammation and fibrosis using biopsy specimens of patients with steatotic liver disease or viral hepatitis. REGISTRATION PROSPERO registration number: CRD42022364598.
Collapse
Affiliation(s)
- Junzhao Ye
- Department of Gastroenterology The First Affiliated Hospital Sun Yat-sen University No. 58 Zhongshan II Road Yuexiu District Guangzhou China
| | - Jiaming Lai
- Department of Gastroenterology The First Affiliated Hospital Sun Yat-sen University No. 58 Zhongshan II Road Yuexiu District Guangzhou China
| | - Ling Luo
- Department of Gastroenterology The First Affiliated Hospital Sun Yat-sen University No. 58 Zhongshan II Road Yuexiu District Guangzhou China
| | - Ting Zhou
- Department of Gastroenterology The First Affiliated Hospital Sun Yat-sen University No. 58 Zhongshan II Road Yuexiu District Guangzhou China
| | - Yanhong Sun
- Department of Laboratory Medicine The First Affiliated Hospital Sun Yat-sen University No. 58 Zhongshan II Road Yuexiu District Guangzhou China.
| | - Bihui Zhong
- Department of Gastroenterology The First Affiliated Hospital Sun Yat-sen University No. 58 Zhongshan II Road Yuexiu District Guangzhou China.
| |
Collapse
|
4
|
Keskin M, Aysegul Arsoy H, Kara O, Sarandol E, Beyaz A, Koca N, Yilmaz Y. Metabolic and Hepatic Profiles of Non-Obese and Obese Metabolic Dysfunction-Associated Steatotic Liver Disease in Adolescents: The Role of FibroScan Parameters,Fibroblast Growth Factor-21, and Cytokeratin-18. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2025; 36:152-161. [PMID: 39910885 PMCID: PMC11900013 DOI: 10.5152/tjg.2025.24760] [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: 12/09/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025]
Abstract
Background/Aims Metabolic dysfunction-associated steatotic liver disease (MASLD) in adolescents, including non-obese phenotypes, is an increasingly important public health issue. The current study investigated the use of controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) as non-invasive tools, along with fibroblast growth factor-21 (FGF-21) and cytokeratin-18 (CK-18), in non-obese MASLD, obese MASLD, and healthy control groups, exploring metabolic and hepatic profiles in these groups. Materials and Methods This cross-sectional study recruited 195 adolescents aged 9-18 years, stratified into controls (n = 92), non-obese MASLD (n = 32), and obese MASLD (n = 39) groups according to FibroScan and MASLD diagnostic criteria. FibroScan measured LSM and CAP, while enzyme-linked immunosorbent assay kit (ELISA) was used to analyze serum FGF-21 and CK-18 levels. Anthropometric, metabolic, and liver enzyme parameters were assessed. Results Metabolic dysfunction-associated steatotic liver disease groups had higher LSM than controls. Fibroblast growth factor-21 levels were significantly higher in MASLD groups, especially in obese MASLD, while CK-18 levels showed variability without significant group differences. Obese MASLD adolescents had marked metabolic dysfunction with higher insulin, homeostasis model assessment for insulin resistance, triglycerides, and liver enzymes compared to non-obese MASLD and controls. Conclusion Fibroblast growth factor-21 has emerged as a potential biomarker for assessing metabolic dysfunction in MASLD, while LSMs from FibroScan provide valuable insights into fibrosis risk. Elevated FGF-21 levels and FibroScan parameters reflect their potential usefulness in non-invasive assessment of MASLD severity, particularly in obese adolescents. However, further longitudinal studies are needed to establish their roles in predicting disease progression and guiding clinical management.
Collapse
Affiliation(s)
- Murat Keskin
- Department of Gastroenterology and Hepatology, KTO Karatay University Faculty of Medicine, Konya, Türkiye
| | - Hanife Aysegul Arsoy
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, University of Health Sciences Bursa, Yüksek İhtisas Training and Research Hospital, Bursa, Türkiye
| | - Ozlem Kara
- Department of Pediatric Endocrinology, University of Health Sciences Bursa, Yüksek İhtisas Training and Research Hospital, Bursa, Türkiye
| | - Emre Sarandol
- Department of Biochemistry, Bursa Uludağ University, Bursa, Türkiye
| | - Aylin Beyaz
- Department of Biochemistry, Bursa Uludağ University, Bursa, Türkiye
| | - Nizameddin Koca
- Department of Internal Medicine, University of Health Sciences Bursa Faculty of Medicine, Bursa City Training and Research Hospital, Bursa, Türkiye
| | - Yusuf Yilmaz
- Department of Gastroenterology and Hepatology, Riza Recep Tayyip Erdoğan University, Rize, Türkiye
| |
Collapse
|
5
|
Schenker RB, Ramirez CB, Jang C, Allayee H, Zhao X, Setchell KDR, Kohli R, Goran MI. Dihydroxyacetone phosphate is a novel predictor of hepatic fibrosis in Latino adolescents with obesity. J Pediatr Gastroenterol Nutr 2025; 80:174-181. [PMID: 39582475 DOI: 10.1002/jpn3.12420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/25/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common pediatric liver disease and can progress to liver fibrosis. Latino adolescents have increased MASLD and fibrosis risk. While fibrosis is diagnosed by biopsy or imaging, more accessible, noninvasive, and economical screening methods are needed. We aimed to use plasma metabolomics/lipidomics to identify potential fibrosis biomarkers in Latino adolescents with obesity. METHODS Liver stiffness (LS) was measured in 93 Latino adolescents with obesity using magnetic resonance elastography. Metabolites and lipids were extracted from plasma and identified on Compound Discoverer. Associations between metabolites/lipids and fibrosis (LS > 2.73 kPa) were determined using linear regression models after covariate adjustment. False discovery rate (FDR) adjusted Pearson's correlations were performed. Analytes yielding significant FDR-adjusted correlations were examined further by receiver operator curve analysis. RESULTS Mean (±standard deviation) alanine transaminase (ALT) was 45.7(±65.2) IU/L, hepatic fat fraction was 12.7(±9.1)%, and LS was 2.4(±0.3) kPa. We identified 795 metabolites and 413 lipids in plasma, but only one single metabolite, dihydroxyacetone phosphate (DHAP), a marker of triglyceride synthesis, was significantly associated with fibrosis after FDR adjustment (p < 0.05). In terms of predicting fibrosis, ALT had an area under the curve (AUC) of 0.79, and DHAP had an AUC of 0.79. When combined, ALT + DHAP had an AUC of 0.89. CONCLUSIONS The combination of ALT + DHAP may have the potential as an accurate, noninvasive test for liver fibrosis. Our data are limited to Latino children with obesity, and a larger cohort should be examined to further validate this novel biomarker.
Collapse
Affiliation(s)
- Rachel B Schenker
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Cuauhtemoc B Ramirez
- Department of Biologic Chemistry, University of California Irvine School of Medicine, Irvine, California, USA
| | - Cholsoon Jang
- Department of Biologic Chemistry, University of California Irvine School of Medicine, Irvine, California, USA
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, California, USA
| | - Xueheng Zhao
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Division of Pathology and Laboratory Medicine, Cincinnati, Ohio, USA
| | - Kenneth D R Setchell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Division of Pathology and Laboratory Medicine, Cincinnati, Ohio, USA
| | - Rohit Kohli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Michael I Goran
- Department of Pediatrics, Division of Endocrinology, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
6
|
Minichmayr IK, Plan EL, Weber B, Ueckert S. A Model-Based Evaluation of Noninvasive Biomarkers to Reflect Histological Nonalcoholic Fatty Liver Disease Scores. Pharm Res 2025; 42:123-135. [PMID: 39702686 PMCID: PMC11785690 DOI: 10.1007/s11095-024-03791-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/24/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) comprises multiple heterogeneous pathophysiological conditions commonly evaluated by suboptimal liver biopsies. This study aimed to elucidate the role of 13 diverse histological liver scores in assessing NAFLD disease activity using an in silico pharmacometric model-based approach. We further sought to investigate various noninvasive patient characteristics for their ability to reflect all 13 histological scores and the NAFLD activity score (NAS). METHODS A histological liver score model was built upon 13 biopsy-based pathological features (binary and categorical scores) from the extensive NASH-CRN (Nonalcoholic Steatohepatitis-Clinical Research Network) observational NAFLD Database study (n = 914 adults) using the concept of item response theory. The impact of 69 noninvasive biomarkers potentially reflecting NAFLD activity was quantitatively described across the entire spectrum of all 13 histological scores. RESULTS The model suggested that four different disease facets underlie the cardinal NAFLD features (steatosis, inflammation, hepatocellular ballooning (= NAS); fibrosis; highest correlations: corrballooning-fibrosis = 0.69/corrinflammation-ballooning = 0.62/corrsteatosis-inflammation = 0.60). The 13 histological liver scores were best described by contrasting noninvasive biomarkers: Age and platelets best reflected the fibrosis score, while alanine and aspartate aminotransferase best described the NAS, with diverging contributions of the three individual NAS components to the results of the overall NAS. CONCLUSIONS An in silico histological liver score model allowed to simultaneously quantitatively analyze 13 features beyond NAS and fibrosis, characterizing different disease facets underlying NAFLD and revealing the contrasting ability of 69 noninvasive biomarkers to reflect the diverse histological (sub-)scores.
Collapse
Affiliation(s)
- Iris K Minichmayr
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Elodie L Plan
- Department of Pharmacy, Uppsala University, Uppsala, Sweden.
- Pharmetheus, Uppsala, Sweden.
| | - Benjamin Weber
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
- Global Translation, Novo Nordisk A/S, Måløv, Denmark
| | - Sebastian Ueckert
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
- Ribocure, Mölndal, Sweden
| |
Collapse
|
7
|
Jorgensen AL, Korver S, Schofield A, Howell L, Clarke JI, Walker LE, Brillant N, Goldring CEP, Pirmohamed M. Establishing reference ranges for circulating biomarkers of drug-induced liver injury in healthy human volunteers. Br J Clin Pharmacol 2024. [PMID: 39676236 DOI: 10.1111/bcp.16371] [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: 06/12/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
AIMS The potential of mechanistic biomarkers to improve prediction of drug-induced liver injury (DILI) and hepatic regeneration is widely acknowledged. We sought to determine reference intervals for new biomarkers of DILI and regeneration, as well as to characterize their natural variability and impact of diurnal variation. METHODS Serum samples from 227 healthy volunteers were recruited as part of a cross-sectional study; of these, 25 subjects had weekly serial sampling over 3 weeks, while 23 had intensive blood sampling over a 24h period. Alanine aminotransferase (ALT), MicroRNA-122 (miR-122), High Mobility Group Box-1 (HMGB1), total Keratin-18 (K18), caspase-cleaved Keratin-18 (ccK18), Glutamate Dehydrogenase (GLDH) and Macrophage Colony-Stimulating Factor-1 (CSF-1) were assayed. RESULTS Reference intervals were established for each biomarker based on the 97.5% quantile (90% CI) following the assessment of fixed effects in univariate and multivariable models. Intra-individual variability was found to be non-significant, and there was no significant impact of diurnal variation. CONCLUSION Reference intervals for novel DILI biomarkers have been described. An upper limit of a reference range might represent the most appropriate mechanism to utilize these data. These data can now be used to interpret data from exploratory clinical DILI studies and to assist their further qualification as required by regulatory authorities.
Collapse
Affiliation(s)
- Andrea L Jorgensen
- Department of Health Data Science, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Samantha Korver
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Amy Schofield
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lawrence Howell
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Joanna I Clarke
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lauren E Walker
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Nathalie Brillant
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Chris E P Goldring
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Munir Pirmohamed
- Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, Molecular & Clinical Pharmacology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
8
|
Park KH, Lee H, Lee JH, Yon DK, Choi YI, Chung HJ, Jung J, Jeong NY. Unique and Shared Molecular Mechanisms of Alcoholic and Non-Alcoholic Liver Cirrhosis Identified Through Transcriptomics Data Integration. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:537-547. [PMID: 39417237 DOI: 10.1089/omi.2024.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Liver cirrhosis is a severe chronic disease that results from various etiological factors and leads to substantial morbidity and mortality. Alcoholic cirrhosis (AC) and non-AC (NAC) arise from prolonged and excessive consumption of alcohol and metabolic syndromes, respectively. Precise molecular mechanisms of AC and NAC are yet to be comprehensively understood for diagnostics and therapeutic advances to materialize. This study reports novel findings to this end by utilizing high-throughput RNA sequencing and microarray data from the Gene Expression Omnibus (GEO). We performed a meta-analysis of transcriptomics data to identify the differentially expressed genes specific to AC and NAC. Functional enrichment and protein-protein interaction network analyses uncovered novel hub genes and transcription factors (TFs) critical to AC and NAC. We found that AC is primarily driven by metabolic dysregulation and oxidative stress, with key TFs such as RELA, NFKB1, and STAT3. NAC was characterized by fibrosis and tissue remodeling associated with metabolic dysfunction, with TFs including USF1, MYCN, and HIF1A. Key hub genes such as ESR1, JUN, FOS, and PKM in AC, and CD8A, MAPK3, CCND1, and CXCR4 in NAC were identified, along with their associated TFs, pointing to potential therapeutic targets. Our results underscore the unique and shared molecular mechanisms that underlie AC and NAC and inform the efforts toward precision medicine and improved patient outcomes in liver cirrhosis.
Collapse
Affiliation(s)
- Ki-Hoon Park
- Department of Anesthesiology and Pain Medicine, College of Medicine, Kosin University, Busan, South Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
| | - Hwajin Lee
- Department of Biomedical Science, Graduation School, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Precision Medicine, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
| | - Ji Hyun Lee
- Department of Biomedical Science, Graduation School, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Precision Medicine, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
| | - Dong Keon Yon
- Department of Precision Medicine, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Digital Health, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
| | - Young-Il Choi
- Department of Surgery, College of Medicine, Kosin University, Busan, South Korea
| | - Hyung-Joo Chung
- Department of Anesthesiology and Pain Medicine, College of Medicine, Kosin University, Busan, South Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Biomedical Science, Graduation School, Kyung Hee University, Dongdaemun-gu, South Korea
- Department of Precision Medicine, College of Medicine, Kyung Hee University, Dongdaemun-gu, South Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan, South Korea
| |
Collapse
|
9
|
Wu Y, Zhou J, Zhang J, Li H. Cytokeratin 18 in nonalcoholic fatty liver disease: value and application. Expert Rev Mol Diagn 2024; 24:1009-1022. [PMID: 39387822 DOI: 10.1080/14737159.2024.2413941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Nonalcoholic fatty liver disease (NAFLD) is a common metabolism-related disease worldwide. Although studies have shown that some medications may be effective for treating NAFLD, they do not satisfy the medical requirements, and lifestyle changes are the most basic strategy. Thus, early detection of NAFLD and timely lifestyle interventions are highly important. AREAS COVERED The traditional diagnostic methods for NAFLD are limited by accuracy, cost, and security issues. Cytokeratin 18 (CK18), which is a marker of apoptosis and overall cell death, is an excellent biomarker for NAFLD. Liver fat accumulation in NAFLD triggers the activation of caspases, which increases the CK18 cleavage and its release into the blood. CK18 can help diagnose different stages of NAFLD, especially the nonalcoholic steatohepatitis (NASH) stage. In evaluating the efficacy of the NAFLD treatment and predicting the risk of NAFLD-related diseases, CK18 plays a significant role. EXPERT OPINION CK18 can non-invasively monitor the pathological conditions of NAFLD patients and provide new hope for the early diagnosis of NAFLD. Adding CK18 to the NAFLD diagnostic criteria that are widely used in clinical settings may be efficient for the detection of NAFLD and early effective intervention.
Collapse
Affiliation(s)
- Yuan Wu
- School of Medicine, The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Jing Zhou
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Jun Zhang
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Hongshan Li
- School of Medicine, The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, China
| |
Collapse
|
10
|
Sezgin Y, Bora ES, Arda DB, Uyanikgil Y, Erbaş O. Caffeine mitigates tamoxifen-induced fatty liver in Wistar rats. Acta Cir Bras 2024; 39:e396924. [PMID: 39356936 PMCID: PMC11441146 DOI: 10.1590/acb396924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Tamoxifen, a widely used drug for breast cancer treatment, is associated with adverse effects on the liver, including the development of fatty liver. This study aimed to investigate the potential protective effect of caffeine against tamoxifen-induced fatty liver in Wistar rats. METHODS Rats were divided into normal control, tamoxifen + saline, and tamoxifen + caffeine. Plasma samples were assessed for biochemical markers related to oxidative stress, inflammation, liver function, and cell damage. Additionally, liver histopathology was examined to quantify the extent of fatty infiltration. RESULTS In the tamoxifen + saline group, elevated levels of plasma malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), alanine aminotransferase (ALT), cytokeratin 18, and soluble ST2 were observed compared to the normal control group, indicating increased oxidative stress, inflammation, and liver injury (p < 0.01). Moreover, histopathological examination revealed a significant increase in fatty infiltration (p < 0.001). However, in the tamoxifen + caffeine group, these markers were markedly reduced (p < 0.05, p < 0.01), and fatty infiltration was significantly mitigated (p < 0.001). CONCLUSIONS The findings suggest that caffeine administration attenuates tamoxifen-induced fatty liver in rats by ameliorating oxidative stress, inflammation, liver injury, and cell damage. Histopathological evidence further supports the protective role of caffeine. This study highlights the potential of caffeine as a therapeutic intervention to counter tamoxifen-induced hepatic complications, contributing to the optimization of breast cancer treatment strategies.
Collapse
Affiliation(s)
- Yasin Sezgin
- Yüzüncü Yıl University - Faculty of Medicine - Clinic of Medical Oncology - Van - Turkey
| | - Ejder Saylav Bora
- İzmir Katip Çelebi University - Faculty of Medicine - Department of Emergency Medicine - Izmir - Turkey
| | - Duygu Burcu Arda
- Ege University - Faculty of Medicine - Department of Histology and Embryology - Izmir - Turkey
| | - Yiğit Uyanikgil
- Taksim Research and Training Hospital - Department of Pediatrics - Istanbul - Turkey
| | - Oytun Erbaş
- Demiroğlu Bilim University - Department of Physiology - Istanbul - Turkey
| |
Collapse
|
11
|
Susman S, Santoso B, Makary MS. Locoregional Therapies for Hepatocellular Carcinoma in Patients with Nonalcoholic Fatty Liver Disease. Biomedicines 2024; 12:2226. [PMID: 39457538 PMCID: PMC11504147 DOI: 10.3390/biomedicines12102226] [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: 09/01/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death worldwide with an average five-year survival rate in the US of 19.6%. With the advent of HBV and HCV treatment and prevention, along with the rising rates of obesity, nonalcoholic fatty liver disease (NAFLD) and metabolic syndrome are set to overtake infectious causes as the most common cause of HCC. While surgical resection and transplantation can be curative when amenable, the disease is most commonly unresectable on presentation, and other treatment approaches are the mainstay of therapy. In these patients, locoregional therapies have evolved as a vital tool in both palliation for advanced disease and as a bridge to surgical resection and transplantation. In this review, we will be exploring the primary locoregional therapies for HCC in patients with NAFLD, including transarterial chemoembolization (TACE), bland transarterial embolization (TAE), transarterial radioembolization (TARE), and percutaneous ablation.
Collapse
Affiliation(s)
- Stephen Susman
- Department of Radiology, Yale University Medical Center, New Haven, CT 06510, USA
| | - Breanna Santoso
- Heritage College of Osteopathic Medicine, Ohio University, Dublin, OH 43016, USA
| | - Mina S. Makary
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43202, USA
| |
Collapse
|
12
|
Binicier OB, Sarı SO, Pakoz ZB, Basok BI. Can serum M30 levels be utilized as an activation marker in patients with ulcerative colitis? REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240704. [PMID: 39292076 PMCID: PMC11404990 DOI: 10.1590/1806-9282.20240704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/17/2024] [Indexed: 09/19/2024]
Abstract
OBJECTIVE Ascertainment of disease activation is an important component of therapeutic decisions in ulcerative colitis patients and may present certain clinical challenges. The objective of this study was to determine serum levels of the M30 fragment of cytokeratin 18 and its utility as an activation marker in patients with ulcerative colitis, who are known to have increased apoptosis. METHODS A total of 60 ulcerative colitis (30 active and 30 remission) patients aged over 18 years and 29 healthy individuals as controls were included in the study. M30, C-reactive protein, and mean platelet volume were evaluated in all participants and compared between ulcerative colitis patients and controls, as well as between those with active disease or remission. RESULTS Although ulcerative colitis patients with active disease had higher M30 levels than those in remission, the difference was not statistically significant (p=0.085). The mean M30 levels tended to increase with increasing extent of involvement, although the differences were not significant (p=0.065). The comparison of C-reactive protein and mean platelet volume according to the site of involvement, however, showed significant differences (p=0.02 and 0.004, respectively). M30 did not show significant correlations with C-reactive protein, mean platelet volume, and Mayo Score (p=0.0834, 0.768, and 0.401, respectively). CONCLUSIONS Our results suggest that, in contrast to C-reactive protein and mean platelet volume, M30 levels do not have a significant role as an activation marker in ulcerative colitis patients. Thus, we believe that M30 may not represent an appropriate marker to be used for this purpose.
Collapse
Affiliation(s)
- Omer Burcak Binicier
- University of Health Sciences Turkey, Izmir Faculty of Medicine, Department of Gastroenterology - İzmir, Turkey
| | - Sevil Ozer Sarı
- University of Health Sciences Turkey, Izmir Faculty of Medicine, Department of Gastroenterology - İzmir, Turkey
| | - Zehra Betul Pakoz
- Katip Celebi University, Ataturk Training and Research Hospital, Department of Gastroenterology - İzmir, Turkey
| | - Banu Isbilen Basok
- University of Health Sciences Turkey, Izmir Faculty of Medicine, Department of Medical Biochemistry - İzmir, Turkey
| |
Collapse
|
13
|
Kurtz JD, Chowdhury SM, Black AK, Lambert AN, Neal AE, Kluthe T, Sparks JD. Cytokeratin-18 is Elevated Prior to Conventional Measures of Liver Disease in Fontan-Associated Liver Disease. Pediatr Cardiol 2024:10.1007/s00246-024-03637-z. [PMID: 39237733 DOI: 10.1007/s00246-024-03637-z] [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: 07/19/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
The Fontan procedure is used to palliate complex forms of congenital heart disease. This results in adverse hepatic sequelae now known as Fontan-associated liver disease (FALD). Conventional laboratory measures of liver disease do not correlate well with FALD severity. Cytokeratin-18 (CK-18) is a measure of cell death and is sensitive in detecting other causes of liver disease. Our aim was to assess the use of a novel measure of liver disease, CK-18, in Fontan patients. This is a single-center, prospective, cross-sectional study of Fontan patients aged 8-21 years old. We performed ultrasound elastography, echocardiography, magnetic resonance imaging, and serum laboratory testing. Novel laboratory test CK-18 levels in Fontan subjects were compared to healthy age-matched controls. Thirteen Fontan patients were evaluated with a median age 15 years (10, 14), 4 Hypoplastic left heart syndrome, 11 were male, and 5 were symptomatic. Fontan patients had normal AST/ALT, but a significantly elevated liver stiffness by elastography (median 13.4 kPa). Hepatic stiffness by elastography was associated with diastolic-indexed (rho = 0.58, p = 0.04) ventricular volumes. Compared to 10 aged-matched controls, CK-18 was higher in the Fontan group-cleaved CK-18 protein (p < 0.01) and full CK-18 protein, (p = 0.02). CK-18 was positively associated with AST and ALT. Elevated CK-18 levels were found in Fontan patients compared to controls suggesting hepatic cell death even in these relatively healthy Fontan patients. CK-18 was elevated prior to changes in traditional testing. CK-18 may be a useful sensitive marker of liver disease in FALD.
Collapse
Affiliation(s)
- Joshua D Kurtz
- Division of Pediatric Cardiology, Department of Pediatrics, University of Louisville, 571 S. Floyd St. Ste 113, Louisville, KY, 40202, USA.
- Norton Children's Hospital Heart Institute, Louisville, KY, USA.
| | - Shahryar M Chowdhury
- Division of Pediatric Cardiology, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Allison K Black
- Division of Pediatric Cardiology, Department of Pediatrics, University of Louisville, 571 S. Floyd St. Ste 113, Louisville, KY, 40202, USA
- Norton Children's Hospital Heart Institute, Louisville, KY, USA
| | - Andrea N Lambert
- Division of Pediatric Cardiology, Department of Pediatrics, University of Louisville, 571 S. Floyd St. Ste 113, Louisville, KY, 40202, USA
- Norton Children's Hospital Heart Institute, Louisville, KY, USA
| | - Ashley E Neal
- Division of Pediatric Cardiology, Department of Pediatrics, University of Louisville, 571 S. Floyd St. Ste 113, Louisville, KY, 40202, USA
- Norton Children's Hospital Heart Institute, Louisville, KY, USA
- Division of Pediatric Cardiology, Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Theresa Kluthe
- Department of Pediatrics, Pediatric Research Institute, University of Louisville, Louisville, KY, USA
| | - Joshua D Sparks
- Division of Pediatric Cardiology, Department of Pediatrics, University of Louisville, 571 S. Floyd St. Ste 113, Louisville, KY, 40202, USA
| |
Collapse
|
14
|
Abdelhameed F, Kite C, Lagojda L, Dallaway A, Chatha KK, Chaggar SS, Dalamaga M, Kassi E, Kyrou I, Randeva HS. Non-invasive Scores and Serum Biomarkers for Fatty Liver in the Era of Metabolic Dysfunction-associated Steatotic Liver Disease (MASLD): A Comprehensive Review From NAFLD to MAFLD and MASLD. Curr Obes Rep 2024; 13:510-531. [PMID: 38809396 PMCID: PMC11306269 DOI: 10.1007/s13679-024-00574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF REVIEW The prevalence of non-alcoholic fatty liver disease (NAFLD) is rapidly increasing worldwide, making it the leading cause of liver related morbidity and mortality. Currently, liver biopsy is the gold standard for assessing individuals with steatohepatitis and fibrosis. However, its invasiveness, sampling variability, and impracticality for large-scale screening has driven the search for non-invasive methods for early diagnosis and staging. In this review, we comprehensively summarise the evidence on the diagnostic performance and limitations of existing non-invasive serum biomarkers and scores in the diagnosis and evaluation of steatosis, steatohepatitis, and fibrosis. RECENT FINDINGS Several non-invasive serum biomarkers and scores have been developed over the last decade, although none has successfully been able to replace liver biopsy. The introduction of new NAFLD terminology, namely metabolic dysfunction-associated fatty liver disease (MAFLD) and more recently metabolic dysfunction-associated steatotic liver disease (MASLD), has initiated a debate on the interchangeability of these terminologies. Indeed, there is a need for more research on the variability of the performance of non-invasive serum biomarkers and scores across the diagnostic entities of NAFLD, MAFLD and MASLD. There remains a significant need for finding valid and reliable non-invasive methods for early diagnosis and assessment of steatohepatitis and fibrosis to facilitate prompt risk stratification and management to prevent disease progression and complications. Further exploration of the landscape of MASLD under the newly defined disease subtypes is warranted, with the need for more robust evidence to support the use of commonly used serum scores against the new MASLD criteria and validation of previously developed scores.
Collapse
Affiliation(s)
- Farah Abdelhameed
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Chris Kite
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- School of Health and Society, Faculty of Education, Health and Wellbeing, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry, CV1 5FB, UK
- Chester Medical School, University of Chester, Shrewsbury, SY3 8HQ, UK
| | - Lukasz Lagojda
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Clinical Evidence-Based Information Service (CEBIS), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Alexander Dallaway
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- School of Health and Society, Faculty of Education, Health and Wellbeing, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | - Kamaljit Kaur Chatha
- Department of Biochemistry and Immunology, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | | | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaupedic and Internal Medicine, Endocrine Unit, Laiko Hospital, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK.
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry, CV1 5FB, UK.
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK.
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
- College of Health, Psychology and Social Care, University of Derby, Derby, DE22 1GB, UK.
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855, Athens, Greece.
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK.
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry, CV1 5FB, UK.
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK.
| |
Collapse
|
15
|
Kawanaka M, Kamada Y, Takahashi H, Iwaki M, Nishino K, Zhao W, Seko Y, Yoneda M, Kubotsu Y, Fujii H, Sumida Y, Kawamoto H, Itoh Y, Nakajima A. Serum Cytokeratin 18 Fragment Is an Indicator for Treating Metabolic Dysfunction-Associated Steatotic Liver Disease. GASTRO HEP ADVANCES 2024; 3:1120-1128. [PMID: 39533978 PMCID: PMC11554606 DOI: 10.1016/j.gastha.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 11/16/2024]
Abstract
Background and Aims Although numerous noninvasive diagnostic methods have been developed to predict liver fibrosis in metabolic dysfunction-associated steatotic liver disease (MASLD), they lack markers for predicting lobular inflammation, hepatocellular ballooning, or changes related to metabolic dysfunction-associated steatohepatitis (MASH). We examined serum cytokeratin 18 fragment (CK18F) as a noninvasive marker for predicting treatment response and "at-risk MASH" and "MASH resolution" in patients with MASLD. Methods One-hundred-and-ten patients with MASLD who underwent repeated biopsy were enrolled (age, 4 [0.5-21] years) in this retrospective study. We investigated associations among serum CK18F levels, liver histology, and blood tests and compared them with changes in serum CK18F levels and liver histology and the resolution of MASH. Additionally, 565 biopsy-proven patients were analyzed for associations among serum CK18F levels, liver histology, and blood tests. Moreover, the Fibrosis-4 (FIB-4) index and CK18F were examined for their usefulness in predicting "at-risk MASH." Results CK18F changes were strongly correlated with changes in lobular inflammation, hepatocellular ballooning, and nonalcoholic fatty liver disease activity score. Multiple regression analysis showed that contributing to "MASH resolution" was associated with changes in CK18F levels as independent factors. Patients diagnosed with MASLD and an FIB-4 index >2.67, or those with an FIB-4 index ≤2.67 and CK18F > 200 U/L, were at high risk of developing MASH and should be referred to a hepatologist. Conversely, those with an FIB-4 index ≤2.67 and CK18F ≤ 200 U/L were effectively managed through regular follow-up appointments. Conclusion CK18F changes are associated with nonalcoholic fatty liver disease activity score changes and are a promising noninvasive diagnostic marker for "at risk MASH" and "MASH resolution."
Collapse
Affiliation(s)
- Miwa Kawanaka
- Department of General Internal Medicine 2, Kawasaki General Medical Center, Kawasaki Medical School, Okayama City, Okayama, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirokazu Takahashi
- Faculty of Medicine, Liver Center, Saga University Hospital, Saga University, Saga, Saga, Japan
- Faculty of Medicine, Division of Metabolism and Endocrinology, Saga University, Saga, Saga, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Ken Nishino
- Department of General Internal Medicine 2, Kawasaki General Medical Center, Kawasaki Medical School, Okayama City, Okayama, Japan
| | - Wenli Zhao
- Faculty of Medicine, Liver Center, Saga University Hospital, Saga University, Saga, Saga, Japan
| | - Yuya Seko
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto Prefecture University of Medicine, Kyoto, Kyoto, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Yoshihito Kubotsu
- Faculty of Medicine, Liver Center, Saga University Hospital, Saga University, Saga, Saga, Japan
| | - Hideki Fujii
- Departments of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Abeno-ku, Osaka, Japan
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare, Tokyo, Japan
| | - Hirofumi Kawamoto
- Department of General Internal Medicine 2, Kawasaki General Medical Center, Kawasaki Medical School, Okayama City, Okayama, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto Prefecture University of Medicine, Kyoto, Kyoto, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, Yokohama, Kanagawa, Japan
| |
Collapse
|
16
|
Kaya S, Boydak M, Aydin M, Aras İ. Association between serum cytokeratin 18 and N-terminal procollagen III propeptide in patients with biopsy-proven nonalcoholic fatty liver disease. Biotech Histochem 2024; 99:313-319. [PMID: 39092622 DOI: 10.1080/10520295.2024.2385011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024] Open
Abstract
Liver biopsy is still the gold standard in the staging of nonalcoholic fatty liver disease (NAFLD), which is the most common chronic liver disease worldwide. However, being an invasive method, liver biopsy has limited use in clinical practice. The aim of this study was to determine the relationship between serum levels of cytokeratin 18 (CK-M30) and N-terminal procollagen III propeptide (PIIINP) in patients with biopsy-proven NAFLD. The study was carried out on volunteers, including both healthy individuals and patients pre-diagnosed with NAFLD. The liver biopsies were re-assessed by applying the Steatosis, Activity, Fibrosis/Fatty Liver Inhibition of Progression (SAF/FLIP) algorithm. At the end of the study, frozen serum samples (-80 °C) were analyzed using commercial kits. CK18-M30 and PIIINP levels significantly differed in all study groups. There was no significant correlation between serum levels of CK18-M30 and PIIINP in healthy individuals but there was a significant positive correlation between CK18-M30 and PIIINP levels in NAFLD (NAFL-nonalcoholic steatohepatitis (NASH)) groups. CK18-M30 was better than PIIINP at distinguishing between NAFL and NASH. The results obtained for biopsy-proven NAFLD demonstrated that both PIIINP and CK18-M30 were partly associated with histological parameters and could aid in distinguishing between NASH and NAFL.
Collapse
Affiliation(s)
- Sercan Kaya
- Health Services Vocational School, Medical Laboratory Program, Batman University, Batman, Turkey
| | - Murat Boydak
- Faculty of Veterinary Medicine Faculty, Department of Histology and Embryology, Selçuk University, Konya, Turkey
| | - Mesut Aydin
- School of Medicine, Department of Gastroenterology, Van Yuzuncu Yil University, Van, Turkey
| | - İbrahim Aras
- School of Medicine, Department of Pathology, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
17
|
Choudhury A, Singh SP, Desmukh A, Sahoo B, Eslam M. Post-Liver Transplant Metabolic Syndrome. J Clin Exp Hepatol 2024; 14:101368. [PMID: 38523736 PMCID: PMC10960134 DOI: 10.1016/j.jceh.2024.101368] [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: 02/09/2023] [Accepted: 02/14/2024] [Indexed: 03/26/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is the second most frequent cause of liver transplantation following alcoholic liver disease. With longer follow-up and increased survival rates, the occurrence rate of the metabolic syndrome is increasing with time among liver transplant recipients. Reappearances of non-alcoholic fatty liver disease after transplantation, both as recurring cases and new instances, are prevalent; nonetheless, the recurrence of fibrosis is minimal. Recognizing populations at elevated risk and enhancing the management of metabolic-related conditions are crucial for maintaining a healthy transplanted organ, particularly considering the prolonged utilization of immunosuppressive treatments. Furthermore, NASH-related cirrhosis patients who had transplant are at a greater risk of cardiovascular, renal events and increased incidence of cancer, necessitating a unique care strategy. This review discusses post-transplant metabolic syndrome, risk factors, pathogenesis, diagnosis, prevention strategy, recurrent and de novo NAFLD and customized immunosuppression.
Collapse
Affiliation(s)
- Ashok Choudhury
- Dept of Hepatology and Liver Transplantation, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Satender P. Singh
- Dept of Hepatology and Liver Transplantation, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Akhil Desmukh
- Dept of Hepatology and Liver Transplantation, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Bishnupriya Sahoo
- Associate Professor of Pediatrics, Consultant Pediatric Gastroenterology, Hepatology and Liver Transplant, SGT University, Gurugram, Haryana, India
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| |
Collapse
|
18
|
Choi SJ, Yoon S, Kim KK, Kim D, Lee HE, Kim KG, Shin SK, Park IB, Kim SM, Lee DH. A Composite Blood Biomarker Including AKR1B10 and Cytokeratin 18 for Progressive Types of Nonalcoholic Fatty Liver Disease. Diabetes Metab J 2024; 48:740-751. [PMID: 38311058 PMCID: PMC11307119 DOI: 10.4093/dmj.2023.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/16/2023] [Indexed: 02/06/2024] Open
Abstract
BACKGRUOUND We aimed to evaluate whether composite blood biomarkers including aldo-keto reductase family 1 member B10 (AKR1B10) and cytokeratin 18 (CK-18; a nonalcoholic steatohepatitis [NASH] marker) have clinically applicable performance for the diagnosis of NASH, advanced liver fibrosis, and high-risk NASH (NASH+significant fibrosis). METHODS A total of 116 subjects including healthy control subjects and patients with biopsy-proven nonalcoholic fatty liver disease (NAFLD) were analyzed to assess composite blood-based and imaging-based biomarkers either singly or in combination. RESULTS A composite blood biomarker comprised of AKR1B10, CK-18, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) showed excellent performance for the diagnosis of, NASH, advanced fibrosis, and high-risk NASH, with area under the receiver operating characteristic curve values of 0.934 (95% confidence interval [CI], 0.888 to 0.981), 0.902 (95% CI, 0.832 to 0.971), and 0.918 (95% CI, 0.862 to 0.974), respectively. However, the performance of this blood composite biomarker was inferior to that various magnetic resonance (MR)-based composite biomarkers, such as proton density fat fraction/MR elastography- liver stiffness measurement (MRE-LSM)/ALT/AST for NASH, MRE-LSM+fibrosis-4 index for advanced fibrosis, and the known MR imaging-AST (MAST) score for high-risk NASH. CONCLUSION Our blood composite biomarker can be useful to distinguish progressive forms of NAFLD as an initial noninvasive test when MR-based tools are not available.
Collapse
Affiliation(s)
- Seung Joon Choi
- Department of Radiology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Sungjin Yoon
- Department of Radiology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Kyoung-Kon Kim
- Department of Family Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Doojin Kim
- Department of Surgery, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Hye Eun Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Kwang Gi Kim
- Department of Biomedical Engineering, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Seung Kak Shin
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Ie Byung Park
- Department of Internal Medicine, 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
|
19
|
Hany M, Demerdash HM, Abouelnasr AA, Torensma B. Effect of Cytokeratin-18, C-peptide, MHR, and MACK-3 Biomarkers in Metabolic Dysfunction-Associated Fatty Liver Disease After Laparoscopic Sleeve Gastrectomy. Biomark Insights 2024; 19:11772719241256496. [PMID: 38836118 PMCID: PMC11149444 DOI: 10.1177/11772719241256496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/05/2024] [Indexed: 06/06/2024] Open
Abstract
Background Laparoscopic sleeve gastrectomy (LSG) has emerged as a valuable treatment for various metabolic disorders, including metabolic dysfunction-associated fatty liver disease (MAFLD) in patients with obesity. Consequently, there is a pressing need to develop noninvasive biomarkers for diagnosing and monitoring disease progression. Objectives This study aimed to evaluate specific biomarkers, including Cytokeratin-18 (CK-18), C-peptide, monocyte to HDL cholesterol ratio (MHR), and MACK-3, in patients with obesity with MAFLD undergoing LSG. Design A prospective cohort study on patients with obesity before and 6 months after the LSG procedure. Methods 70 patients with obesity with confirmed MAFLD, determined by Transient Elastography (TE), were pre- and 6 months postoperatively tested. Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), lipid profile, ghrelin, leptin, peptide YY, GLP-1, and liver fibrosis scores, including AST/ALT ratio (AAR), Fibrosis-4 index (FIB-4), and BARD Score were tested. Results BMI significantly decreased in all participants, with a % excess weight loss of 62.0% ± 15.4%. TE measurements revealed a significant postoperative reduction from 100% to 87.1% (P = .006). All selected biomarkers showed significant postoperative improvement-a significant association of CK-18 with MAFLD markers, including AAR, FIB-4, and BARD score, were found. MACK-3 had positive associations with FIB-4. C-peptide and MHR showed no association with MAFLD markers. Furthermore, there was a positive correlation between CK-18 and MACK-3 tests and between C-peptide and CK-18 and MACK-3. Additionally, a receiver operating characteristic (ROC) curve was constructed, with CK-18 performing the best, with an estimated area under the curve of 0.863. Conclusion Serum CK-18 outperformed other selected biomarkers in predicting and monitoring MAFLD in patients with obesity, suggesting its prospective utility in clinical practice. Further studies are needed to validate the accuracy of the MACK-3 test.
Collapse
Affiliation(s)
- Mohamed Hany
- Department of Surgery, Medical Research Institute, Alexandria University, Alexandria, Egypt
- Madina Women's Hospital, Alexandria, Egypt
| | - Hala M Demerdash
- Department of Clinical Pathology, Alexandria University, Alexandria, Egypt
| | | | - Bart Torensma
- Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
20
|
Solleiro-Villavicencio H, Méndez-García LA, Ocampo-Aguilera NA, Baltazar-Pérez I, Arreola-Miranda JA, Aguayo-Guerrero JA, Alfaro-Cruz A, González-Chávez A, Fonseca-Sánchez MA, Fragoso JM, Escobedo G. Decreased Hepatic and Serum Levels of IL-10 Concur with Increased Lobular Inflammation in Morbidly Obese Patients. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:862. [PMID: 38929479 PMCID: PMC11205754 DOI: 10.3390/medicina60060862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and ranges from simple steatosis to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. Accumulating evidence in animal models suggests that loss of interleukin-10 (IL-10) anti-inflammatory actions might contribute to lobular inflammation, considered one of the first steps toward NASH development. However, the role of IL-10 in lobular inflammation remains poorly explored in humans. We examined mRNA and protein levels of IL-10 in liver biopsies and serum samples from morbidly obese patients, investigating the relationship between IL-10 and lobular inflammation degree. Materials and Methods: We prospectively enrolled morbidly obese patients of both sexes, assessing the lobular inflammation grade by the Brunt scoring system to categorize participants into mild (n = 7), moderate (n = 19), or severe (n = 13) lobular inflammation groups. We quantified the hepatic mRNA expression of IL-10 by quantitative polymerase chain reaction and protein IL-10 levels in liver and serum samples by Luminex Assay. We estimated statistical differences by one-way analysis of variance (ANOVA) and Tukey's multiple comparison test. Results: The hepatic expression of IL-10 significantly diminished in patients with severe lobular inflammation compared with the moderate lobular inflammation group (p = 0.01). The hepatic IL-10 protein levels decreased in patients with moderate or severe lobular inflammation compared with the mild lobular inflammation group (p = 0.008 and p = 0.0008, respectively). In circulation, IL-10 also significantly decreased in subjects with moderate or severe lobular inflammation compared with the mild lobular inflammation group (p = 0.005 and p < 0.0001, respectively). Conclusions: In liver biopsies and serum samples of morbidly obese patients, the protein levels of IL-10 progressively decrease as lobular inflammation increases, supporting the hypothesis that lobular inflammation develops because of the loss of the IL-10-mediated anti-inflammatory counterbalance.
Collapse
Affiliation(s)
| | - Lucía Angélica Méndez-García
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Nydia A. Ocampo-Aguilera
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Itzel Baltazar-Pérez
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - José A. Arreola-Miranda
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - José A. Aguayo-Guerrero
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| | - Ana Alfaro-Cruz
- Pathological Anatomy Department, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | - Antonio González-Chávez
- Clínica de Atención Integral para Pacientes con Diabetes y Obesidad (CAIDO), General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico;
| | | | - José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Sección XVI, Tlalpan, Mexico City 14080, Mexico;
| | - Galileo Escobedo
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga”, Mexico City 06726, Mexico; (L.A.M.-G.); (N.A.O.-A.); (I.B.-P.); (J.A.A.-M.); (J.A.A.-G.)
| |
Collapse
|
21
|
Wang YF, Zhang WL, Li ZX, Liu Y, Tan J, Yin HZ, Zhang ZC, Piao XJ, Ruan MH, Dai ZH, Wang SJ, Mu CY, Yuan JH, Sun SH, Liu H, Yang F. METTL14 downregulation drives S100A4 + monocyte-derived macrophages via MyD88/NF-κB pathway to promote MAFLD progression. Signal Transduct Target Ther 2024; 9:91. [PMID: 38627387 PMCID: PMC11021505 DOI: 10.1038/s41392-024-01797-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Without intervention, a considerable proportion of patients with metabolism-associated fatty liver disease (MAFLD) will progress from simple steatosis to metabolism-associated steatohepatitis (MASH), liver fibrosis, and even hepatocellular carcinoma. However, the molecular mechanisms that control progressive MAFLD have yet to be fully determined. Here, we unraveled that the expression of the N6-methyladenosine (m6A) methyltransferase METTL14 is remarkably downregulated in the livers of both patients and several murine models of MAFLD, whereas hepatocyte-specific depletion of this methyltransferase aggravated lipid accumulation, liver injury, and fibrosis. Conversely, hepatic Mettl14 overexpression alleviated the above pathophysiological changes in mice fed on a high-fat diet (HFD). Notably, in vivo and in vitro mechanistic studies indicated that METTL14 downregulation decreased the level of GLS2 by affecting the translation efficiency mediated by YTHDF1 in an m6A-depedent manner, which might help to form an oxidative stress microenvironment and accordingly recruit Cx3cr1+Ccr2+ monocyte-derived macrophages (Mo-macs). In detail, Cx3cr1+Ccr2+ Mo-macs can be categorized into M1-like macrophages and S100A4-positive macrophages and then further activate hepatic stellate cells (HSCs) to promote liver fibrosis. Further experiments revealed that CX3CR1 can activate the transcription of S100A4 via CX3CR1/MyD88/NF-κB signaling pathway in Cx3cr1+Ccr2+ Mo-macs. Restoration of METTL14 or GLS2, or interfering with this signal transduction pathway such as inhibiting MyD88 could ameliorate liver injuries and fibrosis. Taken together, these findings indicate potential therapies for the treatment of MAFLD progression.
Collapse
Affiliation(s)
- Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, 200438, Shanghai, China
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Wen-Li Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, 200438, Shanghai, China
| | - Zhi-Xuan Li
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, 100048, Beijing, China
| | - Yue Liu
- The Department of Pharmaceutical Analysis, School of Pharmacy, Naval Medical University, 200433, Shanghai, China
| | - Jian Tan
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, 200438, Shanghai, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, 200438, Shanghai, China
| | - Min-Hao Ruan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, 200438, Shanghai, China
| | - Zhi-Hui Dai
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Si-Jie Wang
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Chen-Yang Mu
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Ji-Hang Yuan
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Shu-Han Sun
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, 200438, Shanghai, China.
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, 200433, Shanghai, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, 200433, Shanghai, China.
- Shanghai Key Laboratory of Medical Biodefense, 200433, Shanghai, China.
| |
Collapse
|
22
|
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
|
23
|
Silva-Sperb AS, Moraes HA, Barcelos STA, de Moura BC, Longo L, Michalczuk MT, Cerski CTS, Uribe-Cruz C, da Silveira TR, Álvares-da-Silva MR, Dall’Alba V. Probiotic supplementation for 24 weeks in patients with non-alcoholic steatohepatitis: the PROBILIVER randomized clinical trial. Front Nutr 2024; 11:1362694. [PMID: 38600992 PMCID: PMC11004384 DOI: 10.3389/fnut.2024.1362694] [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: 12/28/2023] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Background and aim Considering the increasing prevalence of non-alcoholic steatohepatitis (NASH) and treatment gaps, this study aimed to evaluate the effect of probiotic supplementation on liver function markers, nutritional status, and clinical parameters. Methods This double-blind, randomized clinical trial (ClinicalTrials.gov ID: NCT0346782) included adult outpatients with biopsy-proven NASH. The intervention consisted of 24 weeks of supplementation with the probiotic mix Lactobacillus acidophilus (1 × 109 CFU) + Lactobacillus rhamnosus (1 × 109 CFU) + Lactobacillus paracasei (1 × 109 CFU) + Bifidobacterium lactis (1 × 109 CFU), or placebo, twice a day. The following parameters were evaluated: demographic and clinical data, transient elastography (FibroScan), liver enzymes, NAFLD fibrosis score, fatty liver index, laboratory assessment, serum concentration of toll-like receptor-4 (sTLR-4) and cytokeratin 18 (CK-18), anthropometric data, dietary intake, and physical activity. Regarding data analysis, the comparison between the groups was based on the delta of the difference of each variable analyzed (value at the end of treatment minus the baseline value) using the t-test for independent samples or the Mann-Whitney U-test. Results Forty-four patients with NASH completed the trial (51.4 ± 11.6 years). At baseline, 87% of participants had a mild liver fibrosis degree on biopsy, normal values of liver enzymes, transient elastography values consistent with grade 1 fibrosis in both groups, increased waist circumference (WC), a BMI of 30.97 kg/m2, and 76% presented with metabolic syndrome (MetS). After the intervention, no differences were observed between the probiotic and placebo groups in terms of MetS, WC, BMI scores, or liver enzyme levels (p > 0.05 for all). The elastography values remained consistent with grade 1 fibrosis in both groups. Although CK-18 was reduced in both groups, a larger effect size was noted in the probiotic group (D = 1.336). sTLR-4 was also reduced in both groups, with no difference between groups (p = 0.885). Conclusion Intervention with probiotics in the early stages of NASH demonstrated no significant change in hepatic and clinical parameters. Clinical trial registration ClinicalTrials.gov, identifier NCT0346782.
Collapse
Affiliation(s)
- Amanda Souza Silva-Sperb
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Helena Abadie Moraes
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Bruna Concheski de Moura
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Larisse Longo
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Matheus Truccolo Michalczuk
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carlos Thadeu Schmidt Cerski
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Unit of Surgical Pathology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carolina Uribe-Cruz
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Themis Reverbel da Silveira
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Mário Reis Álvares-da-Silva
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Valesca Dall’Alba
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Nutrition Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
24
|
Zeng Q, Liu CH, Ampuero J, Wu D, Jiang W, Zhou L, Li H, Bai L, Romero-Gómez M, Tang H. Circular RNAs in non-alcoholic fatty liver disease: Functions and clinical significance. RNA Biol 2024; 21:1-15. [PMID: 38113132 PMCID: PMC10761141 DOI: 10.1080/15476286.2023.2290769] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2023] [Indexed: 12/21/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), which affects approximately 25% of the global population, is an urgent health issue leading to various metabolic comorbidities. Circular RNAs (circRNAs), covalently closed RNA molecules, are characterized by ubiquity, diversity, stability, and conservatism. Indeed, they participate in various biological processes via distinct mechanisms that could modify the natural history of NAFLD. In this review, we briefly introduce the biogenesis, characteristics, and biological functions of circRNAs. Furthermore, we summarize circRNAs expression profiles in NAFLD by intersecting seven sequencing data sets and describe the cellular roles of circRNAs and their potential advantages as biomarkers of NAFLD. In addition, we emphatically discuss the exosomal non-coding RNA sorting mechanisms and possible functions in recipient cells. Finally, we extensively discuss the potential application of targeting disease-related circRNAs and competing endogenous RNA networks through gain-of-function and loss-of-function approaches in targeted therapy of NAFLD.
Collapse
Affiliation(s)
- Qingmin Zeng
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Javier Ampuero
- Digestive Diseases Unit, Virgen del Rocío University Hospital. SeLiver group at Institute of Biomedicine of Seville (IBIS: HUVRocío/CSIC/US). University of Seville, Seville, Spain
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyun Zhou
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Manuel Romero-Gómez
- Digestive Diseases Unit, Virgen del Rocío University Hospital. SeLiver group at Institute of Biomedicine of Seville (IBIS: HUVRocío/CSIC/US). University of Seville, Seville, Spain
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Kazi IN, Kuo L, Tsai E. Noninvasive Methods for Assessing Liver Fibrosis and Steatosis. Gastroenterol Hepatol (N Y) 2024; 20:21-29. [PMID: 38405045 PMCID: PMC10885415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Accurate diagnosis and staging of liver fibrosis is crucial to the individualized management of patients with chronic liver disease. Liver biopsy remains the reference standard for the assessment of steatosis, necroinflammation, and fibrosis. However, over the past decade, there has been an exponential growth in noninvasive tests (NITs) designed to assess liver fibrosis and steatosis. These NITs range from serum biomarkers to imaging assessments of liver tissue stiffness. Current noninvasive methods overcome the limitations of non-specific laboratory markers, conventional imaging, and invasive procedures, and are now starting to be adopted. The Fibrosis-4 index, Enhanced Liver Fibrosis test, and elastography have gained the strongest clinical footholds for the diagnosis of advanced fibrosis. There remains significant interest in demonstrating superiority of any specific test or, alternatively, optimizing a sequential algorithm to provide the most accurate diagnosis of fibrosis staging. This article reviews currently available noninvasive methods for assessing liver fibrosis and steatosis.
Collapse
Affiliation(s)
| | - Lily Kuo
- UT Health San Antonio, San Antonio, Texas
| | - Eugenia Tsai
- UT Health San Antonio, San Antonio, Texas
- Texas Liver Institute, San Antonio, Texas
| |
Collapse
|
26
|
Wegermann K, Moylan C, Naggie S. Fatty Liver Disease: Enter the Metabolic Era. Curr HIV/AIDS Rep 2023; 20:405-418. [PMID: 37882965 DOI: 10.1007/s11904-023-00669-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/27/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to summarize the recent literature linking HIV to metabolic dysfunction-associated steatotic liver disease (MASLD). This is a pressing issue due to the scale of the MASLD epidemic and the urgent need for preventive and therapeutic strategies for MASLD in PWH. RECENT FINDINGS The prevalence of MASLD in PWH is higher than previously appreciated, approaching 50% depending on the population and definition of MASLD. MASLD in PWH is likely multifactorial due to risk factors present in the general population such as metabolic syndrome, and features unique to HIV including systemic inflammation and ART. Statin therapy results in a significant reduction in major adverse cardiovascular events in PWH. PWH are at high risk for MASLD. Screening PWH with metabolic syndrome features could enable earlier interventions to reduce morbidity and mortality associated with MASLD in PWH.
Collapse
Affiliation(s)
- Kara Wegermann
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Cynthia Moylan
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, USA
- Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Susanna Naggie
- Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC, USA.
- Division of Infectious Diseases, Department of Medicine, Duke University Health System, Durham, NC, USA.
| |
Collapse
|
27
|
Liu J, Hu S, Chen L, Daly C, Prada Medina CA, Richardson TG, Traylor M, Dempster NJ, Mbasu R, Monfeuga T, Vujkovic M, Tsao PS, Lynch JA, Voight BF, Chang KM, Million VA, Cobbold JF, Tomlinson JW, van Duijn CM, Howson JMM. Profiling the genome and proteome of metabolic dysfunction-associated steatotic liver disease identifies potential therapeutic targets. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.30.23299247. [PMID: 38076879 PMCID: PMC10705663 DOI: 10.1101/2023.11.30.23299247] [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] [Indexed: 12/21/2023]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) affects over 25% of the population and currently has no effective treatments. Plasma proteins with causal evidence may represent promising drug targets. We aimed to identify plasma proteins in the causal pathway of MASLD and explore their interaction with obesity. METHODS We analysed 2,941 plasma proteins in 43,978 European participants from UK Biobank. We performed genome-wide association study (GWAS) for all MASLD-associated proteins and created the largest MASLD GWAS (109,885 cases/1,014,923 controls). We performed Mendelian Randomization (MR) and integrated proteins and their encoding genes in MASLD ranges to identify candidate causal proteins. We then validated them through independent replication, exome sequencing, liver imaging, bulk and single-cell gene expression, liver biopsies, pathway, and phenome-wide data. We explored the role of obesity by MR and multivariable MR across proteins, body mass index, and MASLD. RESULTS We found 929 proteins associated with MASLD, reported five novel genetic loci associated with MASLD, and identified 17 candidate MASLD protein targets. We identified four novel targets for MASLD (CD33, GRHPR, HMOX2, and SCG3), provided protein evidence supporting roles of AHCY, FCGR2B, ORM1, and RBKS in MASLD, and validated nine previously known targets. We found that CD33, FCGR2B, ORM1, RBKS, and SCG3 mediated the association of obesity and MASLD, and HMOX2, ORM1, and RBKS had effect on MASLD independent of obesity. CONCLUSIONS This study identified new protein targets in the causal pathway of MASLD, providing new insights into the multi-omics architecture and pathophysiology of MASLD. These findings advise further therapeutic interventions for MASLD.
Collapse
Affiliation(s)
- Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Sile Hu
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Lingyan Chen
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Charlotte Daly
- Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | | | - Tom G Richardson
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Matthew Traylor
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Niall J Dempster
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Richard Mbasu
- Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Thomas Monfeuga
- AI & Digital Research, Research & Early Development, Novo Nordisk Research Centre Oxford, UK
| | - Marijana Vujkovic
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Julie A Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, Utah, USA
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Benjamin F Voight
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - V A Million
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
- Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- AI & Digital Research, Research & Early Development, Novo Nordisk Research Centre Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, Utah, USA
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust and the University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Jeremy F Cobbold
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust and the University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust and the University of Oxford, Oxford, UK
| | | | - Joanna M M Howson
- Genetics Centre-of-Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| |
Collapse
|
28
|
Gîlcă-Blanariu GE, Budur DS, Mitrică DE, Gologan E, Timofte O, Bălan GG, Olteanu VA, Ștefănescu G. Advances in Noninvasive Biomarkers for Nonalcoholic Fatty Liver Disease. Metabolites 2023; 13:1115. [PMID: 37999211 PMCID: PMC10672868 DOI: 10.3390/metabo13111115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) currently represents one of the most common liver diseases worldwide. Early diagnosis and disease staging is crucial, since it is mainly asymptomatic, but can progress to nonalcoholic steatohepatitis (NASH) or cirrhosis or even lead to the development of hepatocellular carcinoma. Over time, efforts have been put into developing noninvasive diagnostic and staging methods in order to replace the use of a liver biopsy. The noninvasive methods used include imaging techniques that measure liver stiffness and biological markers, with a focus on serum biomarkers. Due to the impressive complexity of the NAFLD's pathophysiology, biomarkers are able to assay different processes involved, such as apoptosis, fibrogenesis, and inflammation, or even address the genetic background and "omics" technologies. This article reviews not only the currently validated noninvasive methods to investigate NAFLD but also the promising results regarding recently discovered biomarkers, including biomarker panels and the combination of the currently validated evaluation methods and serum markers.
Collapse
Affiliation(s)
- Georgiana-Emmanuela Gîlcă-Blanariu
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Daniela Simona Budur
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
| | - Dana Elena Mitrică
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Elena Gologan
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
| | - Oana Timofte
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Gheorghe Gh Bălan
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Vasile Andrei Olteanu
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| | - Gabriela Ștefănescu
- Gastroenterology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (G.-E.G.-B.); (D.E.M.); (E.G.); (O.T.); (G.G.B.); (V.A.O.)
- Department of Gastroenterology, “Sf Spiridon” County Clinical Emergency Hospital, 100115 Iași, Romania
| |
Collapse
|
29
|
Kakehashi A, Suzuki S, Wanibuchi H. Recent Insights into the Biomarkers, Molecular Targets and Mechanisms of Non-Alcoholic Steatohepatitis-Driven Hepatocarcinogenesis. Cancers (Basel) 2023; 15:4566. [PMID: 37760534 PMCID: PMC10527326 DOI: 10.3390/cancers15184566] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) or metabolic dysfunction-associated steatotic liver disease (MASLD) and steatohepatitis (NASH) are chronic hepatic conditions leading to hepatocellular carcinoma (HCC) development. According to the recent "multiple-parallel-hits hypothesis", NASH could be caused by abnormal metabolism, accumulation of lipids, mitochondrial dysfunction, and oxidative and endoplasmic reticulum stresses and is found in obese and non-obese patients. Recent translational research studies have discovered new proteins and signaling pathways that are involved not only in the development of NAFLD but also in its progression to NASH, cirrhosis, and HCC. Nevertheless, the mechanisms of HCC developing from precancerous lesions have not yet been fully elucidated. Now, it is of particular importance to start research focusing on the discovery of novel molecular pathways that mediate alterations in glucose and lipid metabolism, which leads to the development of liver steatosis. The role of mTOR signaling in NASH progression to HCC has recently attracted attention. The goals of this review are (1) to highlight recent research on novel genetic and protein contributions to NAFLD/NASH; (2) to investigate how recent scientific findings might outline the process that causes NASH-associated HCC; and (3) to explore the reliable biomarkers/targets of NAFLD/NASH-associated hepatocarcinogenesis.
Collapse
Affiliation(s)
- Anna Kakehashi
- Department of Molecular Pathology, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan; (S.S.); (H.W.)
| | | | | |
Collapse
|
30
|
Ugonabo O, Udoh UAS, Rajan PK, Reeves H, Arcand C, Nakafuku Y, Joshi T, Finley R, Pierre SV, Sanabria JR. The Current Status of the Liver Liquid Biopsy in MASH Related HCC: Overview and Future Directions. Biomolecules 2023; 13:1369. [PMID: 37759769 PMCID: PMC10526956 DOI: 10.3390/biom13091369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is one of the major risk factors for chronic liver disease and hepatocellular carcinoma (HCC). The incidence of MASH in Western countries continues to rise, driving HCC as the third cause of cancer-related death worldwide. HCC has become a major global health challenge, partly from the obesity epidemic promoting metabolic cellular disturbances but also from the paucity of biomarkers for its early detection. Over 50% of HCC cases are clinically present at a late stage, where curative measures are no longer beneficial. Currently, there is a paucity of both specific and sensitive biological markers for the early-stage detection of HCC. The search for biological markers in the diagnosis of early HCC in high-risk populations is intense. We described the potential role of surrogates for a liver biopsy in the screening and monitoring of patients at risk for nesting HCC.
Collapse
Affiliation(s)
- Onyinye Ugonabo
- Department of Medicine, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (O.U.); (T.J.)
| | - Utibe-Abasi Sunday Udoh
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Pradeep Kumar Rajan
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Heather Reeves
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Christina Arcand
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Yuto Nakafuku
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Tejas Joshi
- Department of Medicine, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (O.U.); (T.J.)
| | - Rob Finley
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
| | - Juan Ramon Sanabria
- Marshall Institute for Interdisciplinary Research, Marshall University School of Medicine, Huntington, WV 25703, USA; (U.-A.S.U.); (P.K.R.); (Y.N.); (S.V.P.)
- Department of Surgery, Marshall University School of Medicine, Marshall University, Huntington, WV 25701, USA; (H.R.); (C.A.); (R.F.)
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
31
|
Curci R, Bianco A, Franco I, Bonfiglio C, Campanella A, Mirizzi A, Giannuzzi V, Cozzolongo R, Veronese N, Osella AR. Lifestyle Modification: Evaluation of the Effects of Physical Activity and Low-Glycemic-Index Mediterranean Diet on Fibrosis Score. Nutrients 2023; 15:3520. [PMID: 37630711 PMCID: PMC10459797 DOI: 10.3390/nu15163520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Non-Alcoholic Fatty Liver Disease (NAFLD) is one the most prevalent causes of chronic liver disease worldwide. In the absence of an approved drug treatment, lifestyle modification is the first intervention strategy. This study aimed to estimate the main effect of two different physical activity (PA) programs, and a Low-Glycemic-Index Mediterranean Diet (LGIMD), or their combined effect on liver fibrosis parameters in subjects with NAFLD. METHODS Subjects with moderate or severe NAFLD grade of severity (n = 144) were randomly assigned to six intervention arms for three months: LGIMD, PA programs, and their combination. Data were collected at baseline, 45 days, and 90 days. Transient elastography was performed to assess the outcome. RESULTS at 90 days, a statistically significant reduction in kPa was found among subjects following LGMID (-2.85, 95% CI -5.24, -0.45) and those following an LGIMD plus PA1 (-2.37, 95% CI -4.39, -0.35) and LGIMD plus Pa2 (-2.21, 95% CI -4.10, -0.32). The contrast between time 2 and time 1 of the LGIMD plus PA2 treatment showed a statistically significant increase, and vice versa: the contrast between time 3 and time 2 of the same treatment showed a statistically significant reduction. The PA1 and PA2 arms also showed reduced kPa, although the results did not reach statistical significance. CONCLUSIONS The intervention arms, LGIMD, LGIMD+PA1, and LGIMD+PA2, reduced the fibrosis score.
Collapse
Affiliation(s)
- Ritanna Curci
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (R.C.); (A.B.); (I.F.); (C.B.); (A.C.)
| | - Antonella Bianco
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (R.C.); (A.B.); (I.F.); (C.B.); (A.C.)
| | - Isabella Franco
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (R.C.); (A.B.); (I.F.); (C.B.); (A.C.)
| | - Caterina Bonfiglio
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (R.C.); (A.B.); (I.F.); (C.B.); (A.C.)
| | - Angelo Campanella
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (R.C.); (A.B.); (I.F.); (C.B.); (A.C.)
| | | | - Vito Giannuzzi
- Gastroenterology Unit, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (V.G.); (R.C.)
| | - Raffaele Cozzolongo
- Gastroenterology Unit, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (V.G.); (R.C.)
| | - Nicola Veronese
- Geriatric Unit, Department of Medicine, University of Palermo, 90100 Palermo, Italy;
| | - Alberto Ruben Osella
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology—IRCCS “S. de Bellis”, Via Turi, 70013 Castellana Grotte, Italy; (R.C.); (A.B.); (I.F.); (C.B.); (A.C.)
| |
Collapse
|
32
|
Jayasekera D, Hartmann P. Noninvasive biomarkers in pediatric nonalcoholic fatty liver disease. World J Hepatol 2023; 15:609-640. [PMID: 37305367 PMCID: PMC10251277 DOI: 10.4254/wjh.v15.i5.609] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/14/2023] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease worldwide among children and adolescents. It encompasses a spectrum of disease, from its mildest form of isolated steatosis, to nonalcoholic steatohepatitis (NASH) to liver fibrosis and cirrhosis, or end-stage liver disease. The early diagnosis of pediatric NAFLD is crucial in preventing disease progression and in improving outcomes. Currently, liver biopsy is the gold standard for diagnosing NAFLD. However, given its invasive nature, there has been significant interest in developing noninvasive methods that can be used as accurate alternatives. Here, we review noninvasive biomarkers in pediatric NAFLD, focusing primarily on the diagnostic accuracy of various biomarkers as measured by their area under the receiver operating characteristic, sensitivity, and specificity. We examine two major approaches to noninvasive biomarkers in children with NAFLD. First, the biological approach that quantifies serological biomarkers. This includes the study of individual circulating molecules as biomarkers as well as the use of composite algorithms derived from combinations of biomarkers. The second is a more physical approach that examines data measured through imaging techniques as noninvasive biomarkers for pediatric NAFLD. Each of these approaches was applied to children with NAFLD, NASH, and NAFLD with fibrosis. Finally, we suggest possible areas for future research based on current gaps in knowledge.
Collapse
Affiliation(s)
- Dulshan Jayasekera
- Department of Internal Medicine and Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, United States
| | - Phillipp Hartmann
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of California San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
33
|
Vatsalya V, Royer AJ, Jha SK, Parthasarathy R, Tiwari H, Feng W, Ramchandani VA, Kirpich IA, McClain CJ. Drinking and laboratory biomarkers, and nutritional status characterize the clinical presentation of early-stage alcohol-associated liver disease. Adv Clin Chem 2023; 114:83-108. [PMID: 37268335 DOI: 10.1016/bs.acc.2023.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Chronic and heavy alcohol consumption is commonly observed in alcohol use disorder (AUD). AUD often leads to alcohol-associated organ injury, including alcohol-associated liver disease (ALD). Approximately 10-20% of patients with AUD progress to ALD. Progression of ALD from the development phase to more advanced states involve the interplay of several pathways, including nutritional alterations. Multiple pathologic processes have been identified in the progression and severity of ALD. However, there are major gaps in the characterization and understanding of the clinical presentation of early-stage ALD as assessed by clinical markers and laboratory measures. Several Institutions and Universities, including the University of Louisville, in collaboration with the National Institutes of Health, have published a series of manuscripts describing early-stage ALD over the past decade. Here, we comprehensively describe early-stage ALD using the liver injury and drinking history markers, and the laboratory biomarkers (with a focus on nutrition status) that are uniquely involved in the development and progression of early-stage ALD.
Collapse
Affiliation(s)
- Vatsalya Vatsalya
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States; Alcohol Research Center, University of Louisville, Louisville, KY, United States; National Institute on Alcohol Abuse and Alcoholism, NIAAA, NIH, Bethesda, MD, United States; Robley Rex VA Medical Center, Louisville, KY, United States.
| | - Amor J Royer
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Suman Kumar Jha
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Ranganathan Parthasarathy
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Harsh Tiwari
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Wenke Feng
- Alcohol Research Center, University of Louisville, Louisville, KY, United States; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY United States
| | - Vijay A Ramchandani
- National Institute on Alcohol Abuse and Alcoholism, NIAAA, NIH, Bethesda, MD, United States
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States; Alcohol Research Center, University of Louisville, Louisville, KY, United States; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY United States; Department of Microbiology and Immunology, University of Louisville, Louisville KY United States
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States; Alcohol Research Center, University of Louisville, Louisville, KY, United States; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY United States; Robley Rex VA Medical Center, Louisville, KY, United States
| |
Collapse
|
34
|
Golabi P, Shah D, Younossi ZM. How to Identify Advanced Nonalcoholic Fatty Liver Disease in the Primary Care Setting. Semin Liver Dis 2023; 43:142-148. [PMID: 37414024 DOI: 10.1055/s-0043-1770984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects 30 to 40% of the population globally and is increasingly considered the most common liver disease. Patients with type 2 diabetes, obesity, and cardiovascular diseases are at especially increased risk for NAFLD. Although most patients with NAFLD do not have progressive liver disease, some patients progress to cirrhosis, liver cancer, and liver mortality. Given the sheer number of patients with NAFLD, the burden of disease is enormous. Despite this large and increasing burden, identification of NAFLD patients at risk for progressive liver disease in the primary care and diabetology practice settings remains highly suboptimal. In this review, our aim is to summarize a stepwise approach to risk stratify patients with NAFLD which should help practitioners in their management of patients with NAFLD.
Collapse
Affiliation(s)
- Pegah Golabi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia
- Department of Medicine, Center for Liver Disease, Inova Fairfax Medical Campus, Falls Church, Virginia
- Inova Medicine, Inova Health System, Falls Church, Virginia
| | - Dipam Shah
- Department of Medicine, Center for Liver Disease, Inova Fairfax Medical Campus, Falls Church, Virginia
| | - Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, Virginia
- Department of Medicine, Center for Liver Disease, Inova Fairfax Medical Campus, Falls Church, Virginia
- Inova Medicine, Inova Health System, Falls Church, Virginia
| |
Collapse
|
35
|
Alomari M, Rashid MU, Chadalavada P, Ragheb J, Zafar H, Suarez ZK, Khazaaleh S, Gonzalez AJ, Castro FJ. Comparison between metabolic-associated fatty liver disease and nonalcoholic fatty liver disease: From nomenclature to clinical outcomes. World J Hepatol 2023; 15:477-496. [PMID: 37206648 PMCID: PMC10190689 DOI: 10.4254/wjh.v15.i4.477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/04/2023] [Accepted: 03/22/2023] [Indexed: 04/20/2023] Open
Abstract
As a result of the obesity epidemic, Nonalcoholic fatty liver disease (NAFLD) and its complications have increased among millions of people. Consequently, a group of experts recommended changing the term NAFLD to an inclusive terminology more reflective of the underlying pathogenesis; metabolic-associated fatty liver disease (MAFLD). This new term of MAFLD has its own disease epidemiology and clinical outcomes prompting efforts in studying its differences from NAFLD. This article discusses the rationale behind the nomenclature change, the main differences, and its clinical implications.
Collapse
Affiliation(s)
- Mohammad Alomari
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| | - Mamoon Ur Rashid
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| | - Pravallika Chadalavada
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| | - Jonathan Ragheb
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| | - Hammad Zafar
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| | - Zoilo Karim Suarez
- Department of Internal Medicine, Florida Atlantic University Charles E Schmidt College of Medicine, Boca Raton, FL 33431, United States
| | - Shrouq Khazaaleh
- Department of Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH 44126, United States
| | - Adalberto Jose Gonzalez
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| | - Fernando J Castro
- Department of Gastroenterology and Hepatology, Cleveland Clinic Florida, Weston, FL 33331, United States
| |
Collapse
|
36
|
Goralska J, Razny U, Gruca A, Zdzienicka A, Micek A, Dembinska-Kiec A, Solnica B, Malczewska-Malec M. Plasma Cytokeratin-18 Fragment Level Reflects the Metabolic Phenotype in Obesity. Biomolecules 2023; 13:biom13040675. [PMID: 37189422 DOI: 10.3390/biom13040675] [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: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
There is growing interest in the non-invasive identification and monitoring of the outcome of liver damage in obese patients. Plasma cytokeratin-18 (CK-18) fragment levels correlate with the magnitude of hepatocyte apoptosis and have recently been proposed to independently predict the presence of non-alcoholic steatohepatitis (NASH). The aim of the study was to analyze the associations of CK-18 with obesity and related complications: insulin resistance, impaired lipid metabolism and the secretion of hepatokines, adipokines and pro-inflammatory cytokines. The study involved 151 overweight and obese patients (BMI 25-40), without diabetes, dyslipidemia or apparent liver disease. Liver function was assessed based on alanine aminotransferase (ALT), gamma-glutamyl transferase (GGT) and the fatty liver index (FLI). CK-18 M30 plasma levels, FGF-21, FGF-19 and cytokines were determined by ELISA. CK-18 values >150 U/l were accompanied by high ALT, GGT and FLI, insulin resistance, postprandial hypertriglyceridemia, elevated FGF-21 and MCP-1 and decreased adiponectin. ALT activity was the strongest independent factor influencing high CK-18 plasma levels, even after an adjustment for age, sex and BMI [β coefficient (95%CI): 0.40 (0.19-0.61)]. In conclusion, the applied CK-18 cut-off point at 150 U/l allows to distinguish between two metabolic phenotypes in obesity.
Collapse
Affiliation(s)
- Joanna Goralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Urszula Razny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Anna Gruca
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Anna Zdzienicka
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Agnieszka Micek
- Institute of Nursing and Midwifery, Jagiellonian University Medical College; Michałowskiego 12, 31-126 Krakow, Poland
| | - Aldona Dembinska-Kiec
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Bogdan Solnica
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| | - Malgorzata Malczewska-Malec
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Skawinska 8, 31-066 Krakow, Poland
| |
Collapse
|
37
|
Munteanu C, Schwartz B. The Effect of Bioactive Aliment Compounds and Micronutrients on Non-Alcoholic Fatty Liver Disease. Antioxidants (Basel) 2023; 12:antiox12040903. [PMID: 37107278 PMCID: PMC10136128 DOI: 10.3390/antiox12040903] [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: 03/20/2023] [Revised: 03/28/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
In the current review, we focused on identifying aliment compounds and micronutrients, as well as addressed promising bioactive nutrients that may interfere with NAFLD advance and ultimately affect this disease progress. In this regard, we targeted: 1. Potential bioactive nutrients that may interfere with NAFLD, specifically dark chocolate, cocoa butter, and peanut butter which may be involved in decreasing cholesterol concentrations. 2. The role of sweeteners used in coffee and other frequent beverages; in this sense, stevia has proven to be adequate for improving carbohydrate metabolism, liver steatosis, and liver fibrosis. 3. Additional compounds were shown to exert a beneficial action on NAFLD, namely glutathione, soy lecithin, silymarin, Aquamin, and cannabinoids which were shown to lower the serum concentration of triglycerides. 4. The effects of micronutrients, especially vitamins, on NAFLD. Even if most studies demonstrate the beneficial role of vitamins in this pathology, there are exceptions. 5. We provide information regarding the modulation of the activity of some enzymes related to NAFLD and their effect on this disease. We conclude that NAFLD can be prevented or improved by different factors through their involvement in the signaling, genetic, and biochemical pathways that underlie NAFLD. Therefore, exposing this vast knowledge to the public is particularly important.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Betty Schwartz
- The Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| |
Collapse
|
38
|
Nemati A, Nikniaz Z, Mota A. Effects of Resveratrol Supplementation on Nonalcoholic Fatty Liver Disease Management. TOP CLIN NUTR 2023. [DOI: 10.1097/tin.0000000000000314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
39
|
Grzych G, Bernard L, Lestrelin R, Tailleux A, Staels B. [State of the art on the pathophysiology, diagnosis and treatment of non-alcoholic steatohepatitis (NASH)]. ANNALES PHARMACEUTIQUES FRANÇAISES 2023; 81:183-201. [PMID: 36126753 DOI: 10.1016/j.pharma.2022.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022]
Abstract
NAFLD or non-alcoholic fatty liver disease is one of the complications of obesity and diabetes, the prevalence of which is increasing. The causes of the pathology and its development towards its severe form, NASH or non-alcoholic steatohepatitis, are multiple and still poorly understood. Many different pharmacological classes are being tested in clinical trials to treat NASH, but no pharmaceutical treatment is currently on the market. Moreover, the diagnosis of certainty is only possible by liver biopsy and histological analysis, an invasive procedure with high risk for the patient. It is therefore necessary to better understand the natural history of the disease in order to identify therapeutic targets, but also to identify markers for the diagnosis and monitoring of the disease using a blood sample, which will allow an improvement in patient management.
Collapse
Affiliation(s)
- G Grzych
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - L Bernard
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - R Lestrelin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - A Tailleux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - B Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| |
Collapse
|
40
|
Harrison SA, Allen AM, Dubourg J, Noureddin M, Alkhouri N. Challenges and opportunities in NASH drug development. Nat Med 2023; 29:562-573. [PMID: 36894650 DOI: 10.1038/s41591-023-02242-6] [Citation(s) in RCA: 159] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/20/2022] [Indexed: 03/11/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its more severe form, nonalcoholic steatohepatitis (NASH), represent a growing worldwide epidemic and a high unmet medical need, as no licensed drugs have been approved thus far. Currently, histopathological assessment of liver biopsies is mandatory as a primary endpoint for conditional drug approval. This requirement represents one of the main challenges in the field, as there is substantial variability in this invasive histopathological assessment, which leads to dramatically high screen-failure rates in clinical trials. Over the past decades, several non-invasive tests have been developed to correlate with liver histology and, eventually, outcomes to assess disease severity and longitudinal changes non-invasively. However, further data are needed to ensure their endorsement by regulatory authorities as alternatives to histological endpoints in phase 3 trials. This Review describes the challenges of drug development in NAFLD-NASH trials and potential mitigating strategies to move the field forward.
Collapse
Affiliation(s)
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | | | | | - Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Chandler, AZ, USA
| |
Collapse
|
41
|
Bantel H, Schulze-Osthoff K. Non-invasive tests for evaluating treatment response in NAFLD. J Hepatol 2023; 78:e101-e102. [PMID: 36007707 DOI: 10.1016/j.jhep.2022.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Heike Bantel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.
| | | |
Collapse
|
42
|
Zhang H, Rios RS, Boursier J, Anty R, Chan WK, George J, Yilmaz Y, Wong VWS, Fan J, Dufour JF, Papatheodoridis G, Chen L, Schattenberg JM, Shi J, Xu L, Wong GLH, Lange NF, Papatheodoridi M, Mi Y, Zhou Y, Byrne CD, Targher G, Feng G, Zheng M. Hepatocyte apoptosis fragment product cytokeratin-18 M30 level and non-alcoholic steatohepatitis risk diagnosis: an international registry study. Chin Med J (Engl) 2023; 136:341-350. [PMID: 36848175 PMCID: PMC10106257 DOI: 10.1097/cm9.0000000000002603] [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: 09/08/2022] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Liver biopsy for the diagnosis of non-alcoholic steatohepatitis (NASH) is limited by its inherent invasiveness and possible sampling errors. Some studies have shown that cytokeratin-18 (CK-18) concentrations may be useful in diagnosing NASH, but results across studies have been inconsistent. We aimed to identify the utility of CK-18 M30 concentrations as an alternative to liver biopsy for non-invasive identification of NASH. METHODS Individual data were collected from 14 registry centers on patients with biopsy-proven non-alcoholic fatty liver disease (NAFLD), and in all patients, circulating CK-18 M30 levels were measured. Individuals with a NAFLD activity score (NAS) ≥5 with a score of ≥1 for each of steatosis, ballooning, and lobular inflammation were diagnosed as having definite NASH; individuals with a NAS ≤2 and no fibrosis were diagnosed as having non-alcoholic fatty liver (NAFL). RESULTS A total of 2571 participants were screened, and 1008 (153 with NAFL and 855 with NASH) were finally enrolled. Median CK-18 M30 levels were higher in patients with NASH than in those with NAFL (mean difference 177 U/L; standardized mean difference [SMD]: 0.87 [0.69-1.04]). There was an interaction between CK-18 M30 levels and serum alanine aminotransferase, body mass index (BMI), and hypertension ( P < 0.001, P = 0.026 and P = 0.049, respectively). CK-18 M30 levels were positively associated with histological NAS in most centers. The area under the receiver operating characteristics (AUROC) for NASH was 0.750 (95% confidence intervals: 0.714-0.787), and CK-18 M30 at Youden's index maximum was 275.7 U/L. Both sensitivity (55% [52%-59%]) and positive predictive value (59%) were not ideal. CONCLUSION This large multicenter registry study shows that CK-18 M30 measurement in isolation is of limited value for non-invasively diagnosing NASH.
Collapse
Affiliation(s)
- Huai Zhang
- Department of Biostatistics and Medical Record, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of Hepatology, MAFLD Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Rafael S. Rios
- Department of Hepatology, MAFLD Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jerome Boursier
- Service d’Hépato-Gastroentérologie et Oncologie Digestive, Centre Hospitalier Universitaire d’Angers, Angers, France
- Laboratoire HIFIH, UPRES EA3859, SFR ICAT 4208, Université d’Angers, Angers, France
| | - Rodolphe Anty
- Université Côte d’Azur, CHU, INSERM, U1065, C3M, 06204 Nice, France
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Marmara University, Istanbul, Turkey
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Jiangao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Jean-François Dufour
- University Clinic for Visceral Surgery and Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - George Papatheodoridis
- Academic Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital, of Athens “Laiko”, Athens, Greece
| | - Li Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai 200000, China
| | - Jörn M. Schattenberg
- Metabolic Liver Research Program I, Department of Medicine, University Medical Center Mainz, Mainz, Germany
| | - Junping Shi
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310000, China
| | - Liang Xu
- Tianjin Second People's Hospital, Tianjin 300000, China
| | - Grace Lai-Hung Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Naomi F. Lange
- University Clinic for Visceral Surgery and Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Margarita Papatheodoridi
- Academic Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital, of Athens “Laiko”, Athens, Greece
| | - Yuqiang Mi
- Tianjin Second People's Hospital, Tianjin 300000, China
| | - Yujie Zhou
- Department of Hepatology, MAFLD Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai 200000, China
| | - Christopher D. Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton, Southampton General Hospital, Southampton, UK
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Gong Feng
- Xi’an Medical University, Xi’an, Shaanxi 710000, China
| | - Minghua Zheng
- Department of Hepatology, MAFLD Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Institute of Hepatology, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
43
|
Shi YW, Fan JG. Surveillance of the progression and assessment of treatment endpoints for nonalcoholic steatohepatitis. Clin Mol Hepatol 2023; 29:S228-S243. [PMID: 36521452 PMCID: PMC10029951 DOI: 10.3350/cmh.2022.0401] [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: 11/15/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is an aggressive form of nonalcoholic fatty liver disease (NAFLD) characterized by steatosis-associated inflammation and liver injury. Without effective treatment or management, NASH can have life-threatening outcomes. Evaluation and identification of NASH patients at risk for adverse outcomes are therefore important. Key issues in screening NASH patients are the assessment of advanced fibrosis, differentiation of NASH from simple steatosis, and monitoring of dynamic changes during follow-up and treatment. Currently, NASH staging and evaluation of the effectiveness for drugs still rely on pathological diagnosis, despite sample error issues and the subjectivity associated with liver biopsy. Optimizing the pathological assessment of liver biopsy samples and developing noninvasive surrogate methods for accessible, accurate, and safe evaluation are therefore critical. Although noninvasive methods including elastography, serum soluble biomarkers, and combined models have been implemented in the last decade, noninvasive diagnostic measurements are not widely applied in clinical practice. More work remains to be done in establishing cost-effective strategies both for screening for at-risk NASH patients and identifying changes in disease severity. In this review, we summarize the current state of noninvasive methods for detecting steatosis, steatohepatitis, and fibrosis in patients with NASH, and discuss noninvasive assessments for screening at-risk patients with a focus on the characteristics that should be monitored at follow-up.
Collapse
Affiliation(s)
- Yi-Wen Shi
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
44
|
Larrey D, D'Erasmo L, O'Brien S, Arca M. Long-term hepatic safety of lomitapide in homozygous familial hypercholesterolaemia. Liver Int 2023; 43:413-423. [PMID: 36520008 PMCID: PMC10107656 DOI: 10.1111/liv.15497] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Lomitapide is a microsomal triglyceride transfer protein inhibitor for patients with homozygous familial hypercholesterolaemia. Due to its mechanism of action, potential hepatic effects of lomitapide are of clinical interest. This study aimed to determine the long-term hepatic safety of lomitapide. METHODS Data were aggregated from the pivotal phase 3 and extension phase clinical trial with lomitapide (median 5.1 years; serum total bilirubin, transaminases, cytokeratin-18 [CK-18] and enhanced liver fibrosis [ELF] score, fat-soluble vitamins and essential fatty acids), 8-year data from the Lomitapide Observational Worldwide Evaluation Registry (LOWER) and real-world evidence from a cohort of patients treated with lomitapide in Italy (hepatic elastography, and FIB-4 score for hepatic fibrosis). RESULTS In the phase 3 trial and the LOWER registry, any asymptomatic excursions in liver transaminase levels were not associated with elevations in bilirubin, and no Hy's law cases were detected in up to 8 years follow-up. There were no clinically relevant increases among hepatic biomarkers CK-18, CK-18 fragments or ELF score and fat-soluble vitamins and essential fatty acids remained above normal levels. In 34 patients treated in Italy with lomita pide for more than 9 years, elevations in hepatic fat were mild-to-moderate; hepatic stiffness remained normal, and the mean FIB-4 score remained below the fibrosis threshold value of 2.67. CONCLUSIONS These data indicate that the hepatic safety of lomitapide remains favourable with no clinically significant elevations in hepatic biomarkers and hepatic stiffness remained normal for more than 9 years follow-up. PHASE 3 TRIAL: NCT00730236; extension phase: NCT00943306; LOWER: NCT02135705.
Collapse
Affiliation(s)
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine, 'Sapienza' University, Rome, Italy
| | | | - Marcello Arca
- Department of Translational and Precision Medicine, 'Sapienza' University, Rome, Italy
| | | |
Collapse
|
45
|
Im GY. Emerging Biomarkers in Alcohol-associated Hepatitis. J Clin Exp Hepatol 2023; 13:103-115. [PMID: 36647419 PMCID: PMC9840081 DOI: 10.1016/j.jceh.2022.07.246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 01/19/2023] Open
Abstract
Alcohol-associated hepatitis (AH) is a clinical syndrome of jaundice, abdominal pain, and anorexia due to prolonged heavy alcohol intake. AH is associated with changes in gene expression, cytokines, immune response, and the gut microbiome. There are limited biomarkers to diagnose and prognosticate in AH, but several non-invasive biomarkers are emerging. In this review, clinical risk-stratifying algorithms, promising AH biomarkers like cytokeratin-18 fragments, genetic polymorphisms, and microRNAs will be reviewed.
Collapse
Key Words
- AH, Alcohol-associated hepatitis
- ALD, alcohol-associated liver disease
- ASCA, anti–Saccharomyces cerevisiae antibodies
- AUC, area under the curve
- FGF, fibroblast growth factor
- GAHS, Glasgow alcohol-associated hepatitis score
- HCC, hepatocellular carcinoma
- MELD, model for end-stage liver disease
- NASH, non-alcohol-associated steatohepatitis
- PPV, positive predictive value
- PT, prothrombin time
- VCTE, vibration-controlled transient elastography
- alcohol-associated hepatitis
- biomarkers
- cytokines
- miRNAs, MicroRNAs
- microRNA
- microbiome
Collapse
Affiliation(s)
- Gene Y. Im
- Icahn School of Medicine at Mount Sinai, Division of Liver Diseases, Recanati/Miller Transplantation Institute, New York, NY, USA
| |
Collapse
|
46
|
Kamada Y, Nakamura T, Isobe S, Hosono K, Suama Y, Ohtakaki Y, Nauchi A, Yasuda N, Mitsuta S, Miura K, Yamamoto T, Hosono T, Yoshida A, Kawanishi I, Fukushima H, Kinoshita M, Umeda A, Kinoshita Y, Fukami K, Miyawaki T, Fujii H, Yoshida Y, Kawanaka M, Hyogo H, Morishita A, Hayashi H, Tobita H, Tomita K, Ikegami T, Takahashi H, Yoneda M, Jun DW, Sumida Y, Okanoue T, Nakajima A. SWOT analysis of noninvasive tests for diagnosing NAFLD with severe fibrosis: an expert review by the JANIT Forum. J Gastroenterol 2023; 58:79-97. [PMID: 36469127 PMCID: PMC9735102 DOI: 10.1007/s00535-022-01932-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/12/2022] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. Nonalcoholic steatohepatitis (NASH) is an advanced form of NAFLD can progress to liver cirrhosis and hepatocellular carcinoma (HCC). Recently, the prognosis of NAFLD/NASH has been reported to be dependent on liver fibrosis degree. Liver biopsy remains the gold standard, but it has several issues that must be addressed, including its invasiveness, cost, and inter-observer diagnosis variability. To solve these issues, a variety of noninvasive tests (NITs) have been in development for the assessment of NAFLD progression, including blood biomarkers and imaging methods, although the use of NITs varies around the world. The aim of the Japan NASH NIT (JANIT) Forum organized in 2020 is to advance the development of various NITs to assess disease severity and/or response to treatment in NAFLD patients from a scientific perspective through multi-stakeholder dialogue with open innovation, including clinicians with expertise in NAFLD/NASH, companies that develop medical devices and biomarkers, and professionals in the pharmaceutical industry. In addition to conventional NITs, artificial intelligence will soon be deployed in many areas of the NAFLD landscape. To discuss the characteristics of each NIT, we conducted a SWOT (strengths, weaknesses, opportunities, and threats) analysis in this study with the 36 JANIT Forum members (16 physicians and 20 company representatives). Based on this SWOT analysis, the JANIT Forum identified currently available NITs able to accurately select NAFLD patients at high risk of NASH for HCC surveillance/therapeutic intervention and evaluate the effectiveness of therapeutic interventions.
Collapse
Affiliation(s)
- Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Takahiro Nakamura
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd., 2-1-1, Osaki, Shinagawa-Ku, Tokyo, 141-6017 Japan
| | - Satoko Isobe
- FibroScan Division, Integral Corporation, 2-25-2, Kamiosaki, Shinagawa-Ku, Tokyo, 141-0021 Japan
| | - Kumiko Hosono
- Immunology, Hepatology & Dermatology Medical Franchise Dept., Medical Division, Novartis Pharma K.K., 1-23-1, Toranomon, Minato-Ku, Tokyo, 105-6333 Japan
| | - Yukiko Suama
- Medical Information Services, Institute of Immunology Co., Ltd., 1-1-10, Koraku, Bunkyo-Ku, Tokyo, 112-0004 Japan
| | - Yukie Ohtakaki
- Product Development 1St Group, Product Development Dept., Fujirebio Inc., 2-1-1, Nishishinjuku, Shinjuku-Ku, Tokyo, 163-0410 Japan
| | - Arihito Nauchi
- Academic Department, GE Healthcare Japan, 4-7-127, Asahigaoka, Hino, Tokyo, 191-8503 Japan
| | - Naoto Yasuda
- Ultrasound Business Area, Siemens Healthcare KK, 1-11-1, Osaki, Shinagawa-Ku, Tokyo, 141-8644 Japan
| | - Soh Mitsuta
- FibroScan Division, Integral Corporation, 2-25-2, Kamiosaki, Shinagawa-Ku, Tokyo, 141-0021 Japan
| | - Kouichi Miura
- Department of Medicine, Division of Gastroenterology, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, Tochigi, 329-0498 Japan
| | - Takuma Yamamoto
- Cardiovascular and Diabetes, Product Marketing Department, Kowa Company, Ltd., 3-4-10, Nihonbashi Honcho, Chuo-Ku, Tokyo, 103-0023 Japan
| | - Tatsunori Hosono
- Clinical Development & Operations Japan, Nippon Boehringer Ingelheim Co., Ltd., 2-1-1, Osaki, Shinagawa-Ku, Tokyo, 141-6017 Japan
| | - Akihiro Yoshida
- Medical Affairs Department, Kowa Company, Ltd., 3-4-14, Nihonbashi Honcho, Chuo-Ku, Tokyo, 103-8433 Japan
| | - Ippei Kawanishi
- R&D Planning Department, EA Pharma Co., Ltd., 2-1-1, Irifune, Chuo-Ku, Tokyo, 104-0042 Japan
| | - Hideaki Fukushima
- Diagnostics Business Area, Siemens Healthcare Diagnostics KK, 1-11-1, Osaki, Shinagawa-Ku, Tokyo, 141-8673 Japan
| | - Masao Kinoshita
- Marketing Dep. H.U. Frontier, Inc., Shinjuku Mitsui Building, 2-1-1, Nishishinjuku, Shinjuku-Ku, Tokyo, 163-0408 Japan
| | - Atsushi Umeda
- Clinical Development Dept, EA Pharma Co., Ltd., 2-1-1, Irifune, Chuo-Ku, Tokyo, 104-0042 Japan
| | - Yuichi Kinoshita
- Global Drug Development Division, Novartis Pharma KK, 1-23-1, Toranomon, Minato-Ku, Tokyo, 105-6333 Japan
| | - Kana Fukami
- 2Nd Product Planning Dept, 2Nd Product Planning Division, Fujirebio Inc, 2-1-1, Nishishinjuku, Shinjuku-Ku, Tokyo, 163-0410 Japan
| | - Toshio Miyawaki
- Medical Information Services, Institute of Immunology Co., Ltd., 1-1-10, Koraku, Bunkyo-Ku, Tokyo, 112-0004 Japan
| | - Hideki Fujii
- Departments of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahi-Machi, Abeno-Ku, Osaka, Osaka 545-8585 Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, 5-7, Kishibe Shinmachi, Suita, Osaka 564-8567 Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine2, Kawasaki Medical School, Kawasaki Medical Center, 2-6-1, Nakasange, Kita-Ku, Okayama, Okayama 700-8505 Japan
| | - Hideyuki Hyogo
- Department of Gastroenterology, JA Hiroshima Kouseiren General Hospital, 1-3-3, Jigozen, Hatsukaichi, Hiroshima 738-8503 Japan ,Hyogo Life Care Clinic Hiroshima, 6-34-1, Enkobashi-Cho, Minami-Ku, Hiroshima, Hiroshima 732-0823 Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1, Oaza Ikenobe, Miki-Cho, Kita-Gun, Kagawa 761-0793 Japan
| | - Hideki Hayashi
- Department of Gastroenterology and Hepatology, Gifu Municipal Hospital, 7-1, Kashima-Cho, Gifu, Gifu 500-8513 Japan
| | - Hiroshi Tobita
- Division of Hepatology, Shimane University Hospital, 89-1, Enya-Cho, Izumo, Shimane 693-8501 Japan
| | - Kengo Tomita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, 3-2, Namiki, Tokorozawa, Saitama 359-8513 Japan
| | - Tadashi Ikegami
- Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, 3-20-1, Chuo, Ami-Machi, Inashiki-Gun, Ibaraki, 300-0395 Japan
| | - Hirokazu Takahashi
- Liver Center, Faculty of Medicine, Saga University Hospital, Saga University, 5-1-1, Nabeshima, Saga, Saga 849-8501 Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa 236-0004 Japan
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, 04763 Korea
| | - Yoshio Sumida
- Division of Hepatology and Pancreatology, Department of Internal Medicine, Aichi Medical University, 21 Yazako Karimata, Nagakute, Aichi, 480-1195, Japan.
| | - Takeshi Okanoue
- Department of Gastroenterology & Hepatology, Saiseikai Suita Hospital, Osaka, 1-2, Kawazono-Cho, Suita, Osaka 564-0013 Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa 236-0004 Japan
| | | |
Collapse
|
47
|
Kord-Varkaneh H, Salehi-Sahlabadi A, Tinsley GM, Santos HO, Hekmatdoost A. Effects of time-restricted feeding (16/8) combined with a low-sugar diet on the management of non-alcoholic fatty liver disease: A randomized controlled trial. Nutrition 2023; 105:111847. [PMID: 36257081 DOI: 10.1016/j.nut.2022.111847] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/23/2022] [Accepted: 09/05/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVES Emerging studies have employed time-restricted feeding (TRF) and a low-sugar diet alone in the management of non-alcoholic fatty liver disease (NAFLD), but their combination has not been tested. The aim of this study was to investigate the effects of TRF combined with a low-sugar diet on NAFLD parameters, cardiometabolic and inflammatory biomarkers, and body composition in patients with NAFLD. METHODS A 12-wk randomized controlled trial was performed to compare the effects of TRF (16 h fasting/8 h feeding daily [16/8]) plus a low-sugar diet versus a control diet based on traditional meal distribution in patients with NAFLD. Changes in body composition, anthropometric indices, and liver and cardiometabolic markers were investigated. RESULTS TRF 16/8 with a low-sugar diet reduced body fat (26.7 ± 5.4 to 24.2 ± 4.9 kg), body weight (83.8 ± 12.7 to 80.5 ± 12.1 kg), waist circumference (104.59 ± 10.47 to 101.91 ± 7.42 cm), and body mass index (29.1 ± 2.6 to 28 ± 2.7 kg/m2), as well as circulating levels of fasting blood glucose and liver (alanine aminotransferase, 34 ± 13.9 to 21.2 ± 5.4 U/L; aspartate aminotransferase, 26.3 ± 6.2 to 20.50 ± 4 U/L; γ-glutamyl transpeptidase, 33 ± 15 to 23.2 ± 11.1 U/L; fibrosis score, 6.3 ± 1 to 5.2 ± 1.2 kPa; and controlled attenuation parameter, 322.9 ± 34.9 to 270.9 ± 36.2 dB/m), lipids (triacylglycerols, 201.5 ± 35.3 to 133.3 ± 48.7 mg/dL; total cholesterol, 190 ± 36.6 to 157.8 ± 33.6 mg/dL; and low-density lipoprotein cholesterol, 104.6 ± 27.3 to 84 ± 26.3 mg/dL), and inflammatory markers (high-sensitivity C-reactive protein, 3.1 ± 1.1 to 2 ± 0.9 mg/L; and cytokeratin-18, 1.35 ± 0.03 to 1.16 ± 0.03 ng/mL). These results were statistically significant (P < 0.05) compared with the control group. CONCLUSIONS TRF plus a low-sugar diet can reduce adiposity and improve liver, lipid, and inflammatory markers in patients with NAFLD.
Collapse
Affiliation(s)
- Hamed Kord-Varkaneh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ammar Salehi-Sahlabadi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Grant M Tinsley
- Department of Kinesiology & Sport Management, Texas Tech University, Lubbock, Texas, USA
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Litwinowicz K, Waszczuk E, Kuzan A, Bronowicka-Szydełko A, Gostomska-Pampuch K, Naporowski P, Gamian A. Alcoholic Liver Disease Is Associated with Elevated Plasma Levels of Novel Advanced Glycation End-Products: A Preliminary Study. Nutrients 2022; 14:nu14245266. [PMID: 36558425 PMCID: PMC9783524 DOI: 10.3390/nu14245266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Elucidating the biochemical mechanisms associated with the progression of alcoholic liver disease (ALD) to more advanced stages such as alcoholic hepatitis (AH) remains an important clinical and scientific challenge. Several hypotheses point to the involvement of advanced glycation end-products (AGEs) in alcohol-associated liver injuries. Recently, we determined the structure of a synthetic, melibiose-derived AGE (MAGE), which was an analog of the novel AGE subgroup AGE10. The primary objective of our study was to determine whether AGE10 was associated with alcoholic hepatitis. The secondary objective was to provide a diagnostic accuracy of AGE10 in AH. To achieve this objective, we examined the plasma levels of AGE10 in 65 healthy individuals and 65 patients with AH. The AGE10 level was measured using a competitive ELISA. Our study confirmed that patients with AH had significantly higher plasma concentrations of AGE10 compared with healthy controls (184.5 ± 71.1 μg/mL and 123.5 ± 44.9 μg/mL, respectively; p < 0.001). In addition, AGE10 showed an acceptable performance as a diagnostic marker of AH, with an AUC of 0.78. In conclusion, AH was associated with elevated levels of novel advanced glycation end-product AGE10.
Collapse
Affiliation(s)
- Kamil Litwinowicz
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Correspondence:
| | - Ewa Waszczuk
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, 50-566 Wroclaw, Poland
| | - Aleksandra Kuzan
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | | | - Kinga Gostomska-Pampuch
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Piotr Naporowski
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| |
Collapse
|
49
|
Chua D, Low ZS, Cheam GX, Ng AS, Tan NS. Utility of Human Relevant Preclinical Animal Models in Navigating NAFLD to MAFLD Paradigm. Int J Mol Sci 2022; 23:14762. [PMID: 36499091 PMCID: PMC9737809 DOI: 10.3390/ijms232314762] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Fatty liver disease is an emerging contributor to disease burden worldwide. The past decades of work established the heterogeneous nature of non-alcoholic fatty liver disease (NAFLD) etiology and systemic contributions to the pathogenesis of the disease. This called for the proposal of a redefinition in 2020 to that of metabolic dysfunction-associated fatty liver disease (MAFLD) to better reflect the current understanding of the disease. To date, several clinical cohort studies comparing NAFLD and MAFLD hint at the relevancy of the new nomenclature in enriching for patients with more severe hepatic injury and extrahepatic comorbidities. However, the underlying systemic pathogenesis is still not fully understood. Preclinical animal models have been imperative in elucidating key biological mechanisms in various contexts, including intrahepatic disease progression, interorgan crosstalk and systemic dysregulation. Furthermore, they are integral in developing novel therapeutics against MAFLD. However, substantial contextual variabilities exist across different models due to the lack of standardization in several aspects. As such, it is crucial to understand the strengths and weaknesses of existing models to better align them to the human condition. In this review, we consolidate the implications arising from the change in nomenclature and summarize MAFLD pathogenesis. Subsequently, we provide an updated evaluation of existing MAFLD preclinical models in alignment with the new definitions and perspectives to improve their translational relevance.
Collapse
Affiliation(s)
- Damien Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Guo Xiang Cheam
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Aik Seng Ng
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
50
|
Chayanupatkul M, Sawatdee W, Chutaputti A, Tangkijvanich P. The Efficacy of Oligonol in Nonalcoholic Fatty Liver Disease: A Randomized Double-Blinded Placebo-Controlled Trial. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2022; 28:904-908. [PMID: 36074799 DOI: 10.1089/jicm.2021.0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Introduction: Oligonol, an oligomerized-polyphenol from Litchi chinensis extract, has been shown to alleviate metabolic syndrome. The aim of this study was to evaluate the effects of oligonol in patients with nonalcoholic fatty liver disease (NAFLD). Methods: Adult patients with NAFLD defined by magnetic resonance imaging-derived proton density fat fraction (MRI-PDFF) ≥11% were enrolled and then randomly assigned to receive either oligonol or placebo capsules. Primary endpoint was ≥30% reduction in MRI-PDFF at 24 weeks. Secondary outcomes were reductions in bodyweight, waist circumference, alanine transaminase, fasting blood sugar, and lipid profiles at week 24. Results: Forty patients were enrolled (n = 20/group). Primary endpoint was achieved in 20% in the oligonol group and 15% in the placebo group (p = 0.50). The authors found a reduction in MRI-PDFF between weeks 0 and 24 in the oligonol group; however, the change was not different from the placebo group. Secondary outcomes were similar between two groups. Discussion: Oligonol has not shown a significant therapeutic effect in NAFLD. Future studies with a longer duration of therapy might be needed to achieve the primary endpoint. Clinical Trial Registration Number: Thai Clinical Trial Registry identification number: TCTR20200814001.
Collapse
Affiliation(s)
- Maneerat Chayanupatkul
- Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases Research Unit, Department of Physiology, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Waleerat Sawatdee
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Anuchit Chutaputti
- Department of Medicine, Phramongkutklao Hospital, Ratchathewi, Bangkok, Thailand
| | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| |
Collapse
|