101
|
Sanyal AJ, Foucquier J, Younossi ZM, Harrison SA, Newsome PN, Chan WK, Yilmaz Y, De Ledinghen V, Costentin C, Zheng MH, Wai-Sun Wong V, Elkhashab M, Huss RS, Myers RP, Roux M, Labourdette A, Destro M, Fournier-Poizat C, Miette V, Sandrin L, Boursier J. Enhanced diagnosis of advanced fibrosis and cirrhosis in individuals with NAFLD using FibroScan-based Agile scores. J Hepatol 2023; 78:247-259. [PMID: 36375686 PMCID: PMC10170177 DOI: 10.1016/j.jhep.2022.10.034] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/05/2022] [Accepted: 10/20/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND & AIMS Currently available non-invasive tests, including fibrosis-4 index (FIB-4) and liver stiffness measurement (LSM by VCTE), are highly effective at excluding advanced fibrosis (AF) (F ≥3) or cirrhosis in people with non-alcoholic fatty liver disease (NAFLD), but only have moderate ability to rule-in these conditions. Our objective was to develop and validate two new scores (Agile 4 and Agile 3+) to identify cirrhosis or AF, respectively, with optimized positive predictive value and fewer indeterminate results, in individuals with NAFLD attending liver clinics. METHODS This international study included seven adult cohorts with suspected NAFLD who underwent liver biopsy, LSM and blood sampling during routine clinical practice or screening for trials. The population was randomly divided into a training set and an internal validation set, on which the best-fitting logistic regression model was built, and performance and goodness of fit were assessed, respectively. Furthermore, both scores were externally validated on two large cohorts. Cut-offs for high sensitivity and specificity were derived in the training set to rule-out and rule-in cirrhosis or AF and then tested in the validation set and compared to FIB-4 and LSM. RESULTS Each score combined LSM, AST/ALT ratio, platelets, sex and diabetes status, as well as age for Agile 3+. Calibration plots for Agile 4 and Agile 3+ indicated satisfactory to excellent goodness of fit. Agile 4 and Agile 3+ outperformed FIB-4 and LSM in terms of AUROC, percentage of patients with indeterminate results and positive predictive value to rule-in cirrhosis or AF. CONCLUSIONS The two novel non-invasive scores improve identification of cirrhosis or AF among individuals with NAFLD attending liver clinics and reduce the need for liver biopsy in this population. IMPACT AND IMPLICATIONS Non-invasive tests currently used to identify patients with advanced fibrosis or cirrhosis, such as fibrosis-4 index and liver stiffness measurement by vibration-controlled transient elastography, have high negative predictive values but high false positive rates, while results are indeterminate for a large number of cases. This study provides scores that will help the clinician diagnose advanced fibrosis or cirrhosis. These new easy-to-implement scores will help liver specialists to better identify (1) patients who need more intensive follow-up, (2) patients who should be referred for inclusion in therapeutic trials, and (3) which patients should be treated with pharmacological agents when effective therapies are approved.
Collapse
Affiliation(s)
- Arun J Sanyal
- Director, Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, VCU School of Medicine and Chair, Division of Gastroenterology, Hepatology and Nutrition in the Department of Internal Medicine at VCU School of Medicine, Richmond, VA, USA
| | | | | | | | - Philip N Newsome
- National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK & Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turkey; Liver Research Unit, Institute of Gastroenterology, Marmara University, Istanbul, Turkey
| | - Victor De Ledinghen
- Hepatology and Gastroenterology Department, Haut-Lévêque University Hospital, Pessac, France
| | - Charlotte Costentin
- Hepato-gastroenterology & Digestive Oncology Department, Grenoble-Alpes University Hospital, Grenoble, France
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, China; Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Ryan S Huss
- Gilead Sciences, Inc., Foster City, CA, USA; The Liver Company, Palo Alto, CA, USA
| | - Robert P Myers
- Gilead Sciences, Inc., Foster City, CA, USA; The Liver Company, Palo Alto, CA, USA
| | - Marine Roux
- HIFIH Laboratory, UPRES EA3859, SFR 4208, Angers University, Angers, France
| | | | | | | | | | | | - Jérôme Boursier
- HIFIH Laboratory, UPRES EA3859, SFR 4208, Angers University, Angers, France; Hepato-Gastroenterology Department, Angers University Hospital, Angers, France
| |
Collapse
|
102
|
Herrera-Marcos LV, Martínez-Beamonte R, Arnal C, Barranquero C, Puente-Lanzarote JJ, Herrero-Continente T, Lou-Bonafonte JM, Gonzalo-Romeo G, Mocciaro G, Jenkins B, Surra JC, Rodríguez-Yoldi MJ, Burillo JC, Lasheras R, García-Gil A, Güemes A, Koulman A, Osada J. Dietary squalene supplementation decreases triglyceride species and modifies phospholipid lipidomic profile in the liver of a porcine model of non-alcoholic steatohepatitis. J Nutr Biochem 2023; 112:109207. [PMID: 36402249 DOI: 10.1016/j.jnutbio.2022.109207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/07/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
Abstract
Squalene is a key minor component of virgin olive oil, the main source of fat in the Mediterranean diet, and had shown to improve the liver metabolism in rabbits and mice. The present research was carried out to find out whether this effect was conserved in a porcine model of hepatic steatohepatitis and to search for the lipidomic changes involved. The current study revealed that a 0.5% squalene supplementation to a steatotic diet for a month led to hepatic accumulation of squalene and decreased triglyceride content as well as area of hepatic lipid droplets without influencing cholesterol content or fiber areas. However, ballooning score was increased and associated with the hepatic squalene content. Of forty hepatic transcripts related to lipid metabolism and hepatic steatosis, only citrate synthase and a non-coding RNA showed decreased expressions. The hepatic lipidome, assessed by liquid chromatography-mass spectrometry in a platform able to analyze 467 lipids, revealed that squalene supplementation increased ceramide, Cer(36:2), and phosphatidylcholine (PC[32:0], PC[33:0] and PC[34:0]) species and decreased cardiolipin, CL(69:5), and triglyceride (TG[54:2], TG[55:0] and TG[55:2]) species. Plasma levels of interleukin 12p40 increased in pigs receiving the squalene diet. The latter also modified plasma lipidome by increasing TG(58:12) and decreasing non-esterified fatty acid (FA 14:0, FA 16:1 and FA 18:0) species without changes in total NEFA levels. Together this shows that squalene-induced changes in hepatic and plasma lipidomic profiles, non-coding RNA and anti-inflammatory interleukin are suggestive of an alleviation of the disease despite the increase in the ballooning score.
Collapse
Affiliation(s)
- Luis V Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Juan J Puente-Lanzarote
- Servicio de Bioquímica Clínica. Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - José M Lou-Bonafonte
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Gonzalo Gonzalo-Romeo
- Servicio General de Apoyo a la Investigación. División de Experimentación Animal, Universidad de Zaragoza, Zaragoza, Spain
| | - Gabriele Mocciaro
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Huesca, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Juan Carlos Burillo
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Zaragoza, Spain
| | - Roberto Lasheras
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Zaragoza, Spain
| | - Agustín García-Gil
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Antonio Güemes
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
103
|
Yip TCF, Lyu F, Lin H, Li G, Yuen PC, Wong VWS, Wong GLH. Non-invasive biomarkers for liver inflammation in non-alcoholic fatty liver disease: present and future. Clin Mol Hepatol 2023; 29:S171-S183. [PMID: 36503204 PMCID: PMC10029958 DOI: 10.3350/cmh.2022.0426] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation is the key driver of liver fibrosis progression in non-alcoholic fatty liver disease (NAFLD). Unfortunately, it is often challenging to assess inflammation in NAFLD due to its dynamic nature and poor correlation with liver biochemical markers. Liver histology keeps its role as the standard tool, yet it is well-known for substantial sampling, intraobserver, and interobserver variability. Serum proinflammatory cytokines and apoptotic markers, namely cytokeratin-18, are well-studied with reasonable accuracy, whereas serum metabolomics and lipidomics have been adopted in some commercially available diagnostic models. Ultrasound and computed tomography imaging techniques are attractive due to their wide availability; yet their accuracies may not be comparable with magnetic resonance imaging-based tools. Machine learning and deep learning models, be they supervised or unsupervised learning, are promising tools to identify various subtypes of NAFLD, including those with dominating liver inflammation, contributing to sustainable care pathways for NAFLD.
Collapse
Affiliation(s)
- Terry Cheuk-Fung Yip
- Medical Data Analytic Centre, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
| | - Fei Lyu
- Department of Computer Science, Hong Kong Baptist University, Hong Kong, China
| | - Huapeng Lin
- Medical Data Analytic Centre, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
| | - Guanlin Li
- Medical Data Analytic Centre, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
| | - Pong-Chi Yuen
- Department of Computer Science, Hong Kong Baptist University, Hong Kong, China
| | - Vincent Wai-Sun Wong
- Medical Data Analytic Centre, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Lai-Hung Wong
- Medical Data Analytic Centre, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, Prince of Wales Hospital and the University is The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
104
|
Tyshko NV, Nikitin NS, Shestakova SI, Sadykova EO, Trebukh MD, Guseva GV, Trusov NV, Aksenov IV, Golokhvast KS, Tsatsakis A, Tutelyan VA. Systemic Biomarkers and Liver Morphology in Rats during Chronic Low-Dose Toxicant Administration against the Background of Vitamin Deficiency. Bull Exp Biol Med 2023; 174:365-369. [PMID: 36729327 DOI: 10.1007/s10517-023-05709-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Indexed: 02/03/2023]
Abstract
Liver morphology, intensity of apoptosis, and activity of xenobiotic metabolism enzymes were studied in a chronic model experiment in rats receiving a mixture of 6 pesticides against the background of life-long diets with adequate and insufficient supply of water-soluble vitamins. The dose of each pesticide in the mixture did not exceed the acceptable daily intake (1 ADI). It was found that chronic exposure to low doses of anthropogenic toxicants in combination with permanent vitamin deficiency provokes a number of liver changes, such as increased apoptosis activity, cytochrome P450 system depletion, steatosis, and inflammatory infiltration, which is a potential health risk factor.
Collapse
Affiliation(s)
- N V Tyshko
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia.
| | - N S Nikitin
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - S I Shestakova
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - E O Sadykova
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - M D Trebukh
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - G V Guseva
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - N V Trusov
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - I V Aksenov
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
| | - K S Golokhvast
- Siberian Federal Scientific Centre of Agro-BioTechnologies, Russian Academy of Sciences, Krasnoobsk, Novosibirsk region, Russia
| | - A Tsatsakis
- I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - V A Tutelyan
- Federal Research Center of Nutrition and Biotechnology, Moscow, Russia
- I. M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
105
|
Hansen CD, Gram-Kampmann EM, Hansen JK, Hugger MB, Madsen BS, Jensen JM, Olesen S, Torp N, Rasmussen DN, Kjærgaard M, Johansen S, Lindvig KP, Andersen P, Thorhauge KH, Brønd JC, Hermann P, Beck-Nielsen H, Detlefsen S, Hansen T, Højlund K, Thiele MS, Israelsen M, Krag A. Effect of Calorie-Unrestricted Low-Carbohydrate, High-Fat Diet Versus High-Carbohydrate, Low-Fat Diet on Type 2 Diabetes and Nonalcoholic Fatty Liver Disease : A Randomized Controlled Trial. Ann Intern Med 2023; 176:10-21. [PMID: 36508737 DOI: 10.7326/m22-1787] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND It remains unclear if a low-carbohydrate, high-fat (LCHF) diet is a possible treatment strategy for type 2 diabetes mellitus (T2DM), and the effect on nonalcoholic fatty liver disease (NAFLD) has not been investigated. OBJECTIVE To investigate the effect of a calorie-unrestricted LCHF diet, with no intention of weight loss, on T2DM and NAFLD compared with a high-carbohydrate, low-fat (HCLF) diet. DESIGN 6-month randomized controlled trial with a 3-month follow-up. (ClinicalTrials.gov: NCT03068078). SETTING Odense University Hospital in Denmark from November 2016 until June 2020. PARTICIPANTS 165 participants with T2DM. INTERVENTION Two calorie-unrestricted diets: LCHF diet with 50 to 60 energy percent (E%) fat, less than 20E% carbohydrates, and 25E% to 30E% proteins and HCLF diet with 50E% to 60E% carbohydrates, 20E% to 30E% fats, and 20E% to 25E% proteins. MEASUREMENTS Glycemic control, serum lipid levels, metabolic markers, and liver biopsies to assess NAFLD. RESULTS The mean age was 56 years (SD, 10), and 58% were women. Compared with the HCLF diet, participants on the LCHF diet had greater improvements in hemoglobin A1c (mean difference in change, -6.1 mmol/mol [95% CI, -9.2 to -3.0 mmol/mol] or -0.59% [CI, -0.87% to -0.30%]) and lost more weight (mean difference in change, -3.8 kg [CI, -6.2 to -1.4 kg]). Both groups had higher high-density lipoprotein cholesterol and lower triglycerides at 6 months. Changes in low-density lipoprotein cholesterol were less favorable in the LCHF diet group than in the HCLF diet group (mean difference in change, 0.37 mmol/L [CI, 0.17 to 0.58 mmol/L] or 14.3 mg/dL [CI, 6.6 to 22.4 mg/dL]). No statistically significant between-group changes were detected in the assessment of NAFLD. Changes were not sustained at the 9-month follow-up. LIMITATION Open-label trial, self-reported adherence, unintended weight loss, and lack of adjustment for multiple comparisons. CONCLUSION Persons with T2DM on a 6-month, calorie-unrestricted, LCHF diet had greater clinically meaningful improvements in glycemic control and weight compared with those on an HCLF diet, but the changes were not sustained 3 months after intervention. PRIMARY FUNDING SOURCE Novo Nordisk Foundation.
Collapse
Affiliation(s)
- Camilla Dalby Hansen
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Eva-Marie Gram-Kampmann
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark (E.M.G., H.B., K.H.)
| | - Johanne Kragh Hansen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Mie Balle Hugger
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Bjørn Stæhr Madsen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Jane Møller Jensen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Sara Olesen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Nikolaj Torp
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Ditlev Nytoft Rasmussen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Maria Kjærgaard
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Stine Johansen
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Katrine Prier Lindvig
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense SV, Denmark (K.P.L.)
| | - Peter Andersen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Katrine Holtz Thorhauge
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Jan Christian Brønd
- Department of Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark (J.C.B.)
| | - Pernille Hermann
- Department of Medical Endocrinology, Odense University Hospital, Odense, Denmark (P.H.)
| | - Henning Beck-Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark (E.M.G., H.B., K.H.)
| | - Sönke Detlefsen
- Institute of Clinical Research, University of Southern Denmark, and Department of Pathology, Odense University Hospital, Odense, Denmark (S.D.)
| | - Torben Hansen
- Novo Nordisk Foundation, Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark (T.H.)
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark (E.M.G., H.B., K.H.)
| | - Maja Sofie Thiele
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark (C.D.H., M.B.H., N.T., M.K., S.J., K.H.T., M.S.T.)
| | - Mads Israelsen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark (J.K.H., B.S.M., J.M.J., S.O., D.N.R., P.A., M.I.)
| | - Aleksander Krag
- Department of Gastroenterology and Hepatology, Odense University Hospital, and Institute of Clinical Research, University of Southern Denmark, Odense C, Denmark (A.K.)
| |
Collapse
|
106
|
Barboza T, Beaufrère H, Reavill D, Susta L. Morphological features of hepatic lipid changes in bearded dragons ( Pogona vitticeps), and a proposed grading system. Vet Pathol 2023; 60:123-132. [PMID: 36250570 DOI: 10.1177/03009858221128921] [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: 01/31/2023]
Abstract
Hepatic lipidosis is commonly diagnosed in pet bearded dragons (Pogona vitticeps). However, there are no studies detailing the histological features of hepatic lipid changes in this species. This study describes the microscopic features of lipid change and details an associated scoring system. Histologic hepatic sections were retrospectively evaluated from 252 bearded dragons submitted for necropsy. Pathologic assessment was used to develop a grading scheme with 2 qualitative, 1 quantitative, and 6 semi-quantitative microscopic parameters, which were refined based on variability. The final grading system developed for diffuse and panlobular lipid accumulation included 2 semi-quantitative and 1 quantitative categories: percentage of hepatocellular vacuolation, fibrosis, and hepatocellular swelling, respectively. Hepatocellular swelling was indirectly quantified by counting the number of nuclei per unit area. There was a strong positive correlation (P < .001) between the percentage of hepatocellular vacuolation and lipid content, a strong negative correlation (P < .001) between nuclear count and lipid content, and a moderate correlation (P < .001) between fibrosis and lipid content. Each category was given a numerical value ranging from 0 to 4, with the sum of each representing the final grade. Cutoff values stratified microscopic changes into mild (final grade 1-4), moderate (5-7), and severe (≥8). There was strong interrater agreement for assessment of vacuolization, fibrosis, and severity classification and moderate for hepatocellular swelling. This study documents the features of hepatic lipid changes in bearded dragons. Although a cutoff to differentiate pathologic from nonpathologic lipid accumulation could not be estimated, the proposed grading scheme can be used to inform future studies.
Collapse
Affiliation(s)
- Trinita Barboza
- Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Cummings School of Veterinary Medicine at Tufts University, North Grafton, MA
| | | | | | - Leonardo Susta
- Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
107
|
Lazarus JV, Castera L, Mark HE, Allen AM, Adams LA, Anstee QM, Arrese M, Alqahtani SA, Bugianesi E, Colombo M, Cusi K, Hagström H, Loomba R, Romero-Gómez M, Schattenberg JM, Thiele M, Valenti L, Wong VWS, Yilmaz Y, Younossi ZM, Francque SM, Tsochatzis EA. Real-world evidence on non-invasive tests and associated cut-offs used to assess fibrosis in routine clinical practice. JHEP Rep 2023; 5:100596. [PMID: 36644239 PMCID: PMC9832273 DOI: 10.1016/j.jhepr.2022.100596] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 01/18/2023] Open
Abstract
Background & Aims Non-invasive tests (NITs) offer a practical solution for advanced fibrosis identification in non-alcoholic fatty liver disease (NAFLD). Despite increasing implementation, their use is not standardised, which can lead to inconsistent interpretation and risk stratification. We aimed to assess the types of NITs and the corresponding cut-offs used in a range of healthcare settings. Methods A survey was distributed to a convenience sample of liver health experts who participated in a global NAFLD consensus statement. Respondents provided information on the NITs used in their clinic with the corresponding cut-offs and those used in established care pathways in their areas. Results There were 35 respondents from 24 countries, 89% of whom practised in tertiary level settings. A total of 14 different NITs were used, and each respondent reported using at least one (median = 3). Of the respondents, 80% reported using FIB-4 and liver stiffness by vibration-controlled transient elastography (Fibroscan®), followed by the NAFLD fibrosis score (49%). For FIB-4, 71% of respondents used a low cut-off of <1.3 (range <1.0 to <1.45) and 21% reported using age-specific cut-offs. For Fibroscan®, 21% of respondents used a single liver stiffness cut-off: 8 kPa in 50%, while the rest used 7.2 kPa, 7.8 kPa and 8.7 kPa. Among the 63% of respondents who used lower and upper liver stiffness cut-offs, there were variations in both values (<5 to <10 kPa and >7.5 to >20 kPa, respectively). Conclusions The cut-offs used for the same NITs for NAFLD risk stratification vary between clinicians. As cut-offs impact test performance, these findings underscore the heterogeneity in risk-assessment and support the importance of establishing consistent guidelines on the standardised use of NITs in NAFLD management. Lay summary Owing to the high prevalence of non-alcoholic fatty liver disease (NAFLD) in the general population it is important to identify those who have more advanced stages of liver fibrosis, so that they can be properly treated. Non-invasive tests (NITs) provide a practical way to assess fibrosis risk in patients. However, we found that the cut-offs used for the same NITs vary between clinicians. As cut-offs impact test performance, these findings highlight the importance of establishing consistent guidelines on the standardised use of NITs to optimise clinical management of NAFLD.
Collapse
Affiliation(s)
- Jeffrey V. Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Laurent Castera
- Université de Paris, UMR1149 (CRI), Inserm, Paris, France & Service d’Hépatologie, AP-HP, Hôpital Beaujon, Clichy, France
| | - Henry E. Mark
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Leon A. Adams
- Department of Hepatology and Liver Transplant Unit, Sir Charles Gairdner Hospital, Medical School, University of Western Australia, Perth, Australia
| | - Quentin M. Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Marco Arrese
- Department of Gastroenterology, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Saleh A. Alqahtani
- Division of Gastroenterology & Hepatology, Johns Hopkins University, Baltimore, MD, USA
- Organ Transplant Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | | | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| | - Hannes Hagström
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Rohit Loomba
- Department of Medicine, NAFLD Research Center, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Manuel Romero-Gómez
- Virgen del Rocio University Hospital, Institute of Biomedicine of Seville (HUVR/CSIC/US), CIBEREHD, University of Seville, Seville, Spain
| | - Jörn M. Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Mainz, Germany
| | - Maja Thiele
- Liver Research Center, Odense University Hospital and University of Southern Denmark, Odense, Denmark
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
- Liver Research Unit, Institute of Gastroenterology, Marmara University, Istanbul, Turkey
| | | | - Sven M. Francque
- Department of Gastroenterology Hepatology, University Hospital Antwerp & Translational Sciences in Inflammation and Immunology TWI2N, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Emmanuel A. Tsochatzis
- University College London Institute for Liver and Digestive Health, Royal Free Hospital, London, United Kingdom
- Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
108
|
Friemel J, Torres I, Brauneis E, Thörner T, Schäffer AA, Gertz EM, Grob T, Seidl K, Weber A, Ried T, Heselmeyer-Haddad K. Single-cell resolved ploidy and chromosomal aberrations in nonalcoholic steatohepatitis-(NASH) induced hepatocellular carcinoma and its precursor lesions. Sci Rep 2022; 12:22622. [PMID: 36587184 PMCID: PMC9805444 DOI: 10.1038/s41598-022-27173-z] [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: 08/31/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH)-induced hepatocellular carcinoma (HCC) and its precursor, nonalcoholic fatty liver disease (NAFLD) are an unmet health issue due to widespread obesity. We assessed copy number changes of genes associated with hepatocarcinogenesis and oxidative pathways at a single-cell level. Eleven patients with NASH-HCC and 11 patients with NAFLD were included. Eight probes were analyzed using multiplex interphase fluorescence in situ hybridization (miFISH), single-cell imaging and phylogenetic tree modelling: Telomerase reverse transcriptase (TERT), C-Myc (MYC), hepatocyte growth factor receptor tyrosine kinase (MET), tumor protein 53 (TP53), cyclin D1 (CCND1), human epidermal growth factor receptor 2 (HER2), the fragile histidine triad gene (FHIT) and FRA16D oxidoreductase (WWOX). Each NASH-HCC tumor had up to 14 distinct clonal signal patterns indicating multiclonality, which correlated with high tumor grade. Changes frequently observed were TP53 losses, 45%; MYC gains, 36%; WWOX losses, 36%; and HER2 gains, 18%. Whole-genome duplications were frequent (82%) with aberrant tetraploid cells evolving from diploid ancestors. Non-tumorous NAFLD/NASH biopsies did not harbor clonal copy number changes. Fine mapping of NASH-HCC using single-cell multiplex FISH shows that branched tumor evolution involves genome duplication and that multiclonality increases with tumor grade. The loss of oxidoreductase WWOX and HER2 gains could be potentially associated with NASH-induced hepatocellular carcinoma.
Collapse
Affiliation(s)
- Juliane Friemel
- grid.417768.b0000 0004 0483 9129Genetics Branch, CCR, National Cancer Institute, NIH, Bethesda, MD USA ,grid.412004.30000 0004 0478 9977Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland ,grid.5734.50000 0001 0726 5157Department of Pathology, University of Bern, Bern, Switzerland
| | - Irianna Torres
- grid.417768.b0000 0004 0483 9129Genetics Branch, CCR, National Cancer Institute, NIH, Bethesda, MD USA
| | - Elizabeth Brauneis
- grid.417768.b0000 0004 0483 9129Genetics Branch, CCR, National Cancer Institute, NIH, Bethesda, MD USA
| | - Tim Thörner
- grid.417768.b0000 0004 0483 9129Genetics Branch, CCR, National Cancer Institute, NIH, Bethesda, MD USA
| | - Alejandro A. Schäffer
- grid.417768.b0000 0004 0483 9129Cancer Data Science Laboratory, CCR, National Cancer Institute, NIH, Bethesda, MD USA ,grid.280285.50000 0004 0507 7840Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD USA
| | - E. Michael Gertz
- grid.417768.b0000 0004 0483 9129Cancer Data Science Laboratory, CCR, National Cancer Institute, NIH, Bethesda, MD USA ,grid.280285.50000 0004 0507 7840Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD USA
| | - Tobias Grob
- grid.5734.50000 0001 0726 5157Department of Pathology, University of Bern, Bern, Switzerland
| | - Kati Seidl
- grid.412004.30000 0004 0478 9977Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Achim Weber
- grid.412004.30000 0004 0478 9977Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Thomas Ried
- grid.417768.b0000 0004 0483 9129Genetics Branch, CCR, National Cancer Institute, NIH, Bethesda, MD USA
| | - Kerstin Heselmeyer-Haddad
- grid.417768.b0000 0004 0483 9129Genetics Branch, CCR, National Cancer Institute, NIH, Bethesda, MD USA
| |
Collapse
|
109
|
Eriksen PL, Thomsen KL, Hamilton-Dutoit S, Vilstrup DMSH, Sørensen M. Experimental non-alcoholic fatty liver disease causes regional liver functional deficits as measured by the capacity for galactose metabolism while whole liver function is preserved. BMC Gastroenterol 2022; 22:541. [PMID: 36575375 PMCID: PMC9793673 DOI: 10.1186/s12876-022-02574-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/09/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Increasing incidence of non-alcoholic fatty liver disease (NAFLD) calls for improved understanding of how the disease affects metabolic liver function. AIMS To investigate in vivo effects of different NAFLD stages on metabolic liver function, quantified as regional and total capacity for galactose metabolism in a NAFLD model. METHODS Male Sprague Dawley rats were fed a high-fat, high-cholesterol diet for 1 or 12 weeks, modelling early or late NAFLD, respectively. Each NAFLD group (n = 8 each) had a control group on standard chow (n = 8 each). Metabolic liver function was assessed by 2-[18F]fluoro-2-deoxy-D-galactose positron emission tomography; regional galactose metabolism was assessed as standardised uptake value (SUV). Liver tissue was harvested for histology and fat quantification. RESULTS Early NAFLD had median 18% fat by liver volume. Late NAFLD had median 32% fat and varying features of non-alcoholic steatohepatitis (NASH). Median SUV reflecting regional galactose metabolism was reduced in early NAFLD (9.8) and more so in late NAFLD (7.4; p = 0.02), both significantly lower than in controls (12.5). In early NAFLD, lower SUV was quantitatively explained by fat infiltration. In late NAFLD, the SUV decrease was beyond that attributable to fat; probably related to structural NASH features. Total capacity for galactose elimination was intact in both groups, which in late NAFLD was attained by increased fat-free liver mass to 21 g, versus 15 g in early NAFLD and controls (both p ≤ 0.002). CONCLUSION Regional metabolic liver function was compromised in NAFLD by fat infiltration and structural changes. Still, whole liver metabolic function was preserved in late NAFLD by a marked increase in the fat-free liver mass.
Collapse
Affiliation(s)
- Peter Lykke Eriksen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200, Aarhus N, Denmark. .,Department of Internal Medicine, Randers Regional Hospital, Skovlyvej 15, 8930, Randers, Denmark.
| | - Karen Louise Thomsen
- grid.154185.c0000 0004 0512 597XDepartment of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Stephen Hamilton-Dutoit
- grid.154185.c0000 0004 0512 597XDepartment of Pathology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - DMSc Hendrik Vilstrup
- grid.154185.c0000 0004 0512 597XDepartment of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Michael Sørensen
- grid.154185.c0000 0004 0512 597XDepartment of Hepatology and Gastroenterology, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200 Aarhus N, Denmark ,grid.154185.c0000 0004 0512 597XDepartment of Nuclear Medicine & PET, Aarhus University Hospital, Palle Juul Jensens Boulevard 99, 8200 Aarhus N, Denmark ,grid.416838.00000 0004 0646 9184Department of Internal Medicine, Viborg Regional Hospital, Heibergs Alle 5A, 8800 Viborg, Denmark
| |
Collapse
|
110
|
Wang J, Qin T, Sun J, Li S, Cao L, Lu X. Non-invasive methods to evaluate liver fibrosis in patients with non-alcoholic fatty liver disease. Front Physiol 2022; 13:1046497. [PMID: 36589424 PMCID: PMC9794751 DOI: 10.3389/fphys.2022.1046497] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic Fatty Liver Disease (NAFLD) is a chronic liver disease that is strongly related to insulin resistance and metabolic syndrome, and it has become the most common liver disorder in developed countries. NAFLD embraces the full pathological process of three conditions: steatosis, non-alcoholic steatohepatitis, and finally, cirrhosis. As NAFLD progresses, symptoms will become increasingly severe as fibrosis develops. Therefore, evaluating the fibrosis stage is crucial for patients with NAFLD. A liver biopsy is currently considered the gold standard for staging fibrosis. However, due to the limitations of liver biopsy, non-invasive alternatives were extensively studied and validated in patients with NAFLD. The advantages of non-invasive methods include their high safety and convenience compared with other invasive approaches. This review introduces the non-invasive methods, summarizes their benefits and limitations, and assesses their diagnostic performance for NAFLD-induced fibrosis.
Collapse
Affiliation(s)
- Jincheng Wang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Qin
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinyu Sun
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiwu Li
- Liver Disease Center, Qinhuangdao Third Hospital, Qinhuangdao, China
| | - Lihua Cao
- Liver Disease Center, Qinhuangdao Third Hospital, Qinhuangdao, China,*Correspondence: Xiaojie Lu, ; Lihua Cao,
| | - Xiaojie Lu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Xiaojie Lu, ; Lihua Cao,
| |
Collapse
|
111
|
Abulikemu A, Zhao X, Xu H, Li Y, Ma R, Yao Q, Wang J, Sun Z, Li Y, Guo C. Silica nanoparticles aggravated the metabolic associated fatty liver disease through disturbed amino acid and lipid metabolisms-mediated oxidative stress. Redox Biol 2022; 59:102569. [PMID: 36512914 PMCID: PMC9763688 DOI: 10.1016/j.redox.2022.102569] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The metabolic associated fatty liver disease (MAFLD) is a public health challenge, leading to a global increase in chronic liver disease. The respiratory exposure of silica nanoparticles (SiNPs) has revealed to induce hepatotoxicity. However, its role in the pathogenesis and progression of MAFLD was severely under-studied. In this context, the hepatic impacts of SiNPs were investigated in vivo and in vitro through using ApoE-/- mice and free fatty acid (FFA)-treated L02 hepatocytes. Histopathological examinations and biochemical analysis showed SiNPs exposure via intratracheal instillation aggravated hepatic steatosis, lipid vacuolation, inflammatory infiltration and even collagen deposition in ApoE-/- mice, companied with increased hepatic ALT, AST and LDH levels. The enhanced fatty acid synthesis and inhibited fatty acid β-oxidation and lipid efflux may account for the increased hepatic TC/TG by SiNPs. Consistently, SiNPs induced lipid deposition and elevated TC in FFA-treated L02 cells. Further, the activation of hepatic oxidative stress was detected in vivo and in vitro, as evidenced by ROS accumulation, elevated MDA, declined GSH/GSSG and down-regulated Nrf2 signaling. Endoplasmic reticulum (ER) stress was also triggered in response to SiNPs-induced lipid accumulation, as reflecting by the remarkable ER expansion and increased BIP expression. More importantly, an UPLC-MS-based metabolomics analysis revealed that SiNPs disturbed the hepatic metabolic profile in ApoE-/- mice, prominently on amino acids and lipid metabolisms. In particular, the identified differential metabolites were strongly correlated to the activation of oxidative stress and ensuing hepatic TC/TG accumulation and liver injuries, contributing to the progression of liver diseases. Taken together, our study showed SiNPs promoted hepatic steatosis and liver damage, resulting in the aggravation of MAFLD progression. More importantly, the disturbed amino acids and lipid metabolisms-mediated oxidative stress was a key contributor to this phenomenon from a metabolic perspective.
Collapse
Affiliation(s)
- Alimire Abulikemu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xinying Zhao
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Hailin Xu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Yan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Ru Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qing Yao
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Ji Wang
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
112
|
Holmer M, Ekstedt M, Nasr P, Zenlander R, Wester A, Tavaglione F, Romeo S, Kechagias S, Stål P, Hagström H. Effect of common genetic variants on the risk of cirrhosis in non-alcoholic fatty liver disease during 20 years of follow-up. Liver Int 2022; 42:2769-2780. [PMID: 36166317 PMCID: PMC9828463 DOI: 10.1111/liv.15438] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/11/2022] [Accepted: 09/25/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS Several genotypes associate with a worse histopathological profile in patients with non-alcoholic fatty liver disease (NAFLD). Whether genotypes impact long-term outcomes is unclear. We investigated the importance of PNPLA3, TM6SF2, MBOAT7 and GCKR genotype for the development of severe outcomes in NAFLD. METHOD DNA samples were collected from 546 patients with NAFLD. Advanced fibrosis was diagnosed by liver biopsy or elastography. Non-alcoholic steatohepatitis (NASH) was histologically defined. Additionally, 5396 controls matched for age, sex and municipality were identified from population-based registers. Events of severe liver disease and all-cause mortality were collected from national registries. Hazard ratios (HRs) adjusted for age, sex, body mass index and type 2 diabetes were estimated with Cox regression. RESULTS In NAFLD, the G/G genotype of PNPLA3 was associated with a higher prevalence of NASH at baseline (odds ratio [OR] 3.67, 95% CI = 1.66-8.08), but not with advanced fibrosis (OR 1.81, 95% CI = 0.79-4.14). After up to 40 years of follow-up, the PNPLA3 G/G genotype was associated with a higher rate of severe liver disease (adjusted hazard ratio [aHR] 2.27, 95% CI = 1.15-4.47) compared with the C/C variant. NAFLD patients developed cirrhosis at a higher rate than controls (aHR 9.00, 95% CI = 6.85-11.83). The PNPLA3 G/G genotype accentuated this rate (aHR 23.32, 95% = CI 9.14-59.47). Overall mortality was not affected by any genetic variant. CONCLUSION The PNPLA3 G/G genotype is associated with an increased rate of cirrhosis in NAFLD. Our results suggest that assessment of the PNPLA3 genotype is of clinical relevance in patients with NAFLD to individualize monitoring and therapeutic strategies.
Collapse
Affiliation(s)
- Magnus Holmer
- Division of Liver and Pancreatic disease, Department of Upper GIKarolinska University HospitalStockholmSweden,Department of Medicine, HuddingeKarolinska InstitutetStockholmSweden
| | - Mattias Ekstedt
- Department of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring SciencesLinköping UniversityLinköpingSweden
| | - Patrik Nasr
- Department of Medicine, HuddingeKarolinska InstitutetStockholmSweden,Department of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring SciencesLinköping UniversityLinköpingSweden
| | - Robin Zenlander
- Department of Medicine, HuddingeKarolinska InstitutetStockholmSweden
| | - Axel Wester
- Department of Medicine, HuddingeKarolinska InstitutetStockholmSweden
| | - Federica Tavaglione
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg LaboratoryUniversity of GothenburgGothenburgSweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg LaboratoryUniversity of GothenburgGothenburgSweden,Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Stergios Kechagias
- Department of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring SciencesLinköping UniversityLinköpingSweden
| | - Per Stål
- Division of Liver and Pancreatic disease, Department of Upper GIKarolinska University HospitalStockholmSweden,Department of Medicine, HuddingeKarolinska InstitutetStockholmSweden
| | - Hannes Hagström
- Division of Liver and Pancreatic disease, Department of Upper GIKarolinska University HospitalStockholmSweden,Department of Medicine, HuddingeKarolinska InstitutetStockholmSweden,Clinical Epidemiology Unit, Department of Medicine, SolnaKarolinska InstitutetStockholmSweden
| |
Collapse
|
113
|
Wear KA, Han A, Rubin JM, Gao J, Lavarello R, Cloutier G, Bamber J, Tuthill T. US Backscatter for Liver Fat Quantification: An AIUM-RSNA QIBA Pulse-Echo Quantitative Ultrasound Initiative. Radiology 2022; 305:526-537. [PMID: 36255312 DOI: 10.1148/radiol.220606] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is believed to affect one-third of American adults. Noninvasive methods that enable detection and monitoring of NAFLD have the potential for great public health benefits. Because of its low cost, portability, and noninvasiveness, US is an attractive alternative to both biopsy and MRI in the assessment of liver steatosis. NAFLD is qualitatively associated with enhanced B-mode US echogenicity, but visual measures of B-mode echogenicity are negatively affected by interobserver variability. Alternatively, quantitative backscatter parameters, including the hepatorenal index and backscatter coefficient, are being investigated with the goal of improving US-based characterization of NAFLD. The American Institute of Ultrasound in Medicine and Radiological Society of North America Quantitative Imaging Biomarkers Alliance are working to standardize US acquisition protocols and data analysis methods to improve the diagnostic performance of the backscatter coefficient in liver fat assessment. This review article explains the science and clinical evidence underlying backscatter for liver fat assessment. Recommendations for data collection are discussed, with the aim of minimizing potential confounding effects associated with technical and biologic variables.
Collapse
Affiliation(s)
- Keith A Wear
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Aiguo Han
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Jonathan M Rubin
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Jing Gao
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Roberto Lavarello
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Guy Cloutier
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Jeffrey Bamber
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| | - Theresa Tuthill
- From the Center for Devices and Radiological Health, U.S. Food and Drug Administration, 10903 New Hampshire Ave, WO62, Room 2114, Silver Spring, MD 20993 (K.A.W.); Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Ill (A.H.); Department of Radiology, University of Michigan, Ann Arbor, Mich (J.M.R.); Ultrasound Research and Education, Rocky Vista University, Ivins, Utah (J.G.); Department of Engineering, Pontificia Universidad Católica del Perú, Lima, Peru (R.L.); Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montreal, Canada (G.C.); Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Division of Radiotherapy and Imaging, Joint Department of Physics, London, UK (J.B.); and Pfizer, Cambridge, Mass (T.T.)
| |
Collapse
|
114
|
Associations between subcutaneous adipocyte hypertrophy and nonalcoholic fatty liver disease. Sci Rep 2022; 12:20519. [PMID: 36443373 PMCID: PMC9705525 DOI: 10.1038/s41598-022-24482-1] [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: 05/17/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
Adipocyte hypertrophy and expression of adipokines in subcutaneous adipose tissue (SAT) have been linked to steatosis, nonalcoholic steatohepatitis (NASH) and fibrosis in morbidly obese (BMI ≥ 40 kg/m2) subjects. It is unknown if this is also true for subjects with NAFLD with lesser degrees of obesity (BMI < 35 kg/m2). Thirty-two subjects with biopsy-proven NAFLD and 15 non-diabetic controls matched for BMI underwent fine-needle biopsies of SAT. Adipocyte volume was calculated. RNA-sequencing of SAT was performed in a subset of 20 NAFLD patients. Adipocyte volume and gene expression levels were correlated to the presence of NASH or significant fibrosis. Subjects with NAFLD had larger adipocyte volume compared with controls, (1939 pL, 95% CI 1130-1662 vs. 854 pL, 95% CI 781-926, p < 0.001). There was no association between adipocyte volume and the presence of NASH. Gene expression of adipokines previously described to correlate with NASH in morbid obesity, was not associated with NASH or fibrosis. Our results suggest that persons with NAFLD have larger SAT adipocytes compared with controls and that adipocytes are involved in the pathophysiology of hepatic steatosis in NAFLD. However, adipocyte volume was not associated with NASH or fibrosis in NAFLD subjects with varying degrees of obesity.
Collapse
|
115
|
Naoumov NV, Brees D, Loeffler J, Chng E, Ren Y, Lopez P, Tai D, Lamle S, Sanyal AJ. Digital pathology with artificial intelligence analyses provides greater insights into treatment-induced fibrosis regression in NASH. J Hepatol 2022; 77:1399-1409. [PMID: 35779659 DOI: 10.1016/j.jhep.2022.06.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 05/21/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Liver fibrosis is a key prognostic determinant for clinical outcomes in non-alcoholic steatohepatitis (NASH). Current scoring systems have limitations, especially in assessing fibrosis regression. Second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) microscopy with artificial intelligence analyses provides standardized evaluation of NASH features, especially liver fibrosis and collagen fiber quantitation on a continuous scale. This approach was applied to gain in-depth understanding of fibrosis dynamics after treatment with tropifexor (TXR), a non-bile acid farnesoid X receptor agonist in patients participating in the FLIGHT-FXR study (NCT02855164). METHOD Unstained sections from 198 liver biopsies (paired: baseline and end-of-treatment) from 99 patients with NASH (fibrosis stage F2 or F3) who received placebo (n = 34), TXR 140 μg (n = 37), or TXR 200 μg (n = 28) for 48 weeks were examined. Liver fibrosis (qFibrosis®), hepatic fat (qSteatosis®), and ballooned hepatocytes (qBallooning®) were quantitated using SHG/TPEF microscopy. Changes in septa morphology, collagen fiber parameters, and zonal distribution within liver lobules were also quantitatively assessed. RESULTS Digital analyses revealed treatment-associated reductions in overall liver fibrosis (qFibrosis®), unlike conventional microscopy, as well as marked regression in perisinusoidal fibrosis in patients who had either F2 or F3 fibrosis at baseline. Concomitant zonal quantitation of fibrosis and steatosis revealed that patients with greater qSteatosis reduction also have the greatest reduction in perisinusoidal fibrosis. Regressive changes in septa morphology and reduction in septa parameters were observed almost exclusively in F3 patients, who were adjudged as 'unchanged' with conventional scoring. CONCLUSION Fibrosis regression following hepatic fat reduction occurs initially in the perisinusoidal regions, around areas of steatosis reduction. Digital pathology provides new insights into treatment-induced fibrosis regression in NASH, which are not captured by current staging systems. LAY SUMMARY The degree of liver fibrosis (tissue scarring) in non-alcoholic steatohepatitis (NASH) is the main predictor of negative clinical outcomes. Accurate assessment of the quantity and architecture of liver fibrosis is fundamental for patient enrolment in NASH clinical trials and for determining treatment efficacy. Using digital microscopy with artificial intelligence analyses, the present study demonstrates that this novel approach has greater sensitivity in demonstrating treatment-induced reversal of fibrosis in the liver than current systems. Furthermore, additional details are obtained regarding the pathogenesis of NASH disease and the effects of therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Arun J Sanyal
- Virginia Commonwealth University School of Medicine, Richmond, United States
| |
Collapse
|
116
|
Chang X, Bian H, Xia M, Zhu X, Sun X, Yang X, Gao J, Lin H, Yan H, Gao X. Postprandial glucose is correlated with an increasing risk of liver fibrosis in Chinese patients with nonalcoholic fatty liver disease. DIABETES & METABOLISM 2022; 48:101377. [PMID: 35858659 DOI: 10.1016/j.diabet.2022.101377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
AIM Type 2 diabetes (T2DM) is closely related to nonalcoholic fatty liver disease (NAFLD) and is an important risk factor for the progression of liver fibrosis, but the role of 2-h postprandial blood glucose (PPG) as a biomarker in this process remains unclear. This study was designed to investigate the relationship between PPG and liver fibrosis in Chinese NAFLD populations with or without T2DM. METHODS This study included three independent NAFLD populations: 1) 618 inpatients with T2DM or pre-diabetes, 2) 255 patients with T2DM or pre-diabetes who underwent liver biopsy, and 3) a prospective community-based cohort without diabetes who completed a median of 4.22 years follow-up. The degree of liver fibrosis was assessed by liver fibrosis stage in subjects with a liver biopsy, and by NAFLD fibrosis score (NFS) in subjects without liver biopsy. RESULTS In the first population, PPG {OR 0.02, [95% CI (0.01-0.03)], P< 0.001} was positively correlated with NFS. In the second population, an increasing PPG was associated with increase in the proportion of advanced liver fibrosis (P = 0.012). Multivariate line regression revealed that PPG {OR 0.03 [95% CI (0.00-0.06)], P = 0.049}was positively associated with liver fibrosis stages. In the third population, PPG {OR 0.103, [95% CI (0.011-0.194) P = 0.028} at baseline was positively associated with NFS at follow-up. Furthermore, changes in PPG were significantly associated with NFS change after follow-up. We did not find a similar association between fasting glucose or HbA1c and liver fibrosis. CONCLUSIONS PPG was independently associated with the severity of liver fibrosis in the Chinese NAFLD population.
Collapse
Affiliation(s)
- Xinxia Chang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Xiaopeng Zhu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Xiaoyang Sun
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Xinyu Yang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| | - Jian Gao
- Center of Clinical Epidemiology and Evidence-based Medicine, Fudan University, Shanghai 200032, China; Department of Nutrition, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China.
| | - Hongmei Yan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China.
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Fudan Institute for Metabolic Diseases, Fudan University, Shanghai 200032, China
| |
Collapse
|
117
|
Lei L, Bruneau A, El Mourabit H, Guégan J, Folseraas T, Lemoinne S, Karlsen TH, Hoareau B, Morichon R, Gonzalez-Sanchez E, Goumard C, Ratziu V, Charbord P, Gautheron J, Tacke F, Jaffredo T, Cadoret A, Housset C. Portal fibroblasts with mesenchymal stem cell features form a reservoir of proliferative myofibroblasts in liver fibrosis. Hepatology 2022; 76:1360-1375. [PMID: 35278227 DOI: 10.1002/hep.32456] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS In liver fibrosis, myofibroblasts derive from HSCs and as yet undefined mesenchymal cells. We aimed to identify portal mesenchymal progenitors of myofibroblasts. APPROACH AND RESULTS Portal mesenchymal cells were isolated from mouse bilio-vascular tree and analyzed by single-cell RNA-sequencing. Thereby, we uncovered the landscape of portal mesenchymal cells in homeostatic mouse liver. Trajectory analysis enabled inferring a small cell population further defined by surface markers used to isolate it. This population consisted of portal fibroblasts with mesenchymal stem cell features (PMSCs), i.e., high clonogenicity and trilineage differentiation potential, that generated proliferative myofibroblasts, contrasting with nonproliferative HSC-derived myofibroblasts (-MF). Using bulk RNA-sequencing, we built oligogene signatures of the two cell populations that remained discriminant across myofibroblastic differentiation. SLIT2, a prototypical gene of PMSC/PMSC-MF signature, mediated profibrotic and angiogenic effects of these cells, which conditioned medium promoted HSC survival and endothelial cell tubulogenesis. Using PMSC/PMSC-MF 7-gene signature and slit guidance ligand 2 fluorescent in situ hybridization, we showed that PMSCs display a perivascular portal distribution in homeostatic liver and largely expand with fibrosis progression, contributing to the myofibroblast populations that form fibrotic septa, preferentially along neovessels, in murine and human liver disorders, irrespective of etiology. We also unraveled a 6-gene expression signature of HSCs/HSC-MFs that did not vary in these disorders, consistent with their low proliferation rate. CONCLUSIONS PMSCs form a small reservoir of expansive myofibroblasts, which, in interaction with neovessels and HSC-MFs that mainly arise through differentiation from a preexisting pool, underlie the formation of fibrotic septa in all types of liver diseases.
Collapse
Affiliation(s)
- Lin Lei
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Haquima El Mourabit
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France
| | - Justine Guégan
- Institut du Cerveau (ICM), Bioinformatics/Biostatistics iCONICS Facility, Sorbonne Université, INSERM, Paris, France
| | - Trine Folseraas
- Division of Surgery, Inflammatory Medicine and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norwegian PSC Research Center, Oslo, Norway
| | - Sara Lemoinne
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Department of Hepatology, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (CRMR MIVB-H, ERN RARE-LIVER), Assistance Publique-Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, Paris, France
| | - Tom Hemming Karlsen
- Division of Surgery, Inflammatory Medicine and Transplantation, Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norwegian PSC Research Center, Oslo, Norway
| | - Bénédicte Hoareau
- Sorbonne Université, INSERM, UMS Production et Analyse de Données en Sciences de la Vie et en Santé (PASS), Cytométrie Pitié-Salpêtrière (CyPS), Paris, France
| | - Romain Morichon
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France
| | - Ester Gonzalez-Sanchez
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France
| | - Claire Goumard
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Departments of Hepatology, Hepatobiliary Surgery and Liver Transplantation, AP-HP, Sorbonne Université, ICAN, Pitié-Salpêtrière Hospital, Paris, France
| | - Vlad Ratziu
- Departments of Hepatology, Hepatobiliary Surgery and Liver Transplantation, AP-HP, Sorbonne Université, ICAN, Pitié-Salpêtrière Hospital, Paris, France
| | - Pierre Charbord
- Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, Sorbonne Université, CNRS, INSERM, Paris, France
| | - Jérémie Gautheron
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, Berlin, Germany
| | - Thierry Jaffredo
- Institut de Biologie Paris Seine (IBPS), Laboratoire de Biologie du Développement, Sorbonne Université, CNRS, INSERM, Paris, France
| | - Axelle Cadoret
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France
| | - Chantal Housset
- Centre de Recherche Saint-Antoine (CRSA), Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Department of Hepatology, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (CRMR MIVB-H, ERN RARE-LIVER), Assistance Publique-Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, Paris, France
| |
Collapse
|
118
|
Ericson E, Bergenholm L, Andréasson A, Dix CI, Knöchel J, Hansson SF, Lee R, Schumi J, Antonsson M, Fjellström O, Nasr P, Liljeblad M, Carlsson B, Kechagias S, Lindén D, Ekstedt M. Hepatic patatin-like phospholipase domain-containing 3 levels are increased in I148M risk allele carriers and correlate with NAFLD in humans. Hepatol Commun 2022; 6:2689-2701. [PMID: 35833455 PMCID: PMC9512469 DOI: 10.1002/hep4.2032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/29/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022] Open
Abstract
In nonalcoholic fatty liver disease (NAFLD) the patatin-like phospholipase domain-containing 3 (PNPLA3) rs738409 variant is a contributor. In mice, the Pnpla3 148M variant accumulates on lipid droplets and probably leads to sequestration of a lipase cofactor leading to impaired mobilization of triglycerides. To advance our understanding of the localization and abundance of PNPLA3 protein in humans, we used liver biopsies from patients with NAFLD to investigate the link to NAFLD and the PNPLA3 148M genotype. We experimentally qualified an antibody against human PNPLA3. Hepatic PNPLA3 protein fractional area and localization were determined by immunohistochemistry in biopsies from a well-characterized NAFLD cohort of 67 patients. Potential differences in hepatic PNPLA3 protein levels among patients related to degree of steatosis, lobular inflammation, ballooning, and fibrosis, and PNPLA3 I148M gene variants were assessed. Immunohistochemistry staining in biopsies from patients with NAFLD showed that hepatic PNPLA3 protein was predominantly localized to the membranes of small and large lipid droplets in hepatocytes. PNPLA3 protein levels correlated strongly with steatosis grade (p = 0.000027) and were also significantly higher in patients with lobular inflammation (p = 0.009), ballooning (p = 0.022), and significant fibrosis (stage 2-4, p = 0.014). In addition, PNPLA3 levels were higher in PNPLA3 rs738409 148M (CG, GG) risk allele carriers compared to 148I (CC) nonrisk allele carriers (p = 0.0029). Conclusion: PNPLA3 protein levels were associated with increased hepatic lipid content and disease severity in patients with NAFLD and were higher in PNPLA3 rs738409 (148M) risk allele carriers. Our hypothesis that increased hepatic levels of PNPLA3 may be part of the pathophysiological mechanism of NAFLD is supported.
Collapse
Affiliation(s)
- Elke Ericson
- Genome EngineeringDiscovery SciencesBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Linnéa Bergenholm
- Drug Metabolism and PharmacokineticsResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Anne‐Christine Andréasson
- Bioscience CardiovascularResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Carly I. Dix
- Discovery BiologyDiscovery Sciences, BioPharmaceuticals R&DAstraZenecaCambridgeUK
| | - Jane Knöchel
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology and Safety SciencesBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Sara F. Hansson
- Translational Science and Experimental MedicineResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Richard Lee
- Antisense Drug DiscoveryIonis PharmaceuticalsCarlsbadCaliforniaUSA
- Preclinical Pharmacology and Translational Medicine, Verve TherapeuticsCambridgeMassachusettsUSA
| | - Jennifer Schumi
- Early Biometrics and Statistical InnovationData Science and Artificial IntelligenceBioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Madeleine Antonsson
- Drug Metabolism and PharmacokineticsResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Ola Fjellström
- ProjectsCardiovascular, Renal, and MetabolismBiopharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Patrik Nasr
- Department of Gastroenterology and HepatologyDepartment of HealthMedicine and Caring SciencesLinköping UniversityLinköpingSweden
| | - Mathias Liljeblad
- Translational Science and Experimental MedicineResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Björn Carlsson
- Early Clinical DevelopmentResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
| | - Stergios Kechagias
- Department of Gastroenterology and HepatologyDepartment of HealthMedicine and Caring SciencesLinköping UniversityLinköpingSweden
| | - Daniel Lindén
- Bioscience MetabolismResearch and Early DevelopmentCardiovascular, Renal, and MetabolismBioPharmaceuticals R&DAstraZenecaGothenburgSweden
- Division of EndocrinologyDepartment of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Mattias Ekstedt
- Department of Gastroenterology and HepatologyDepartment of HealthMedicine and Caring SciencesLinköping UniversityLinköpingSweden
| |
Collapse
|
119
|
Ivashkin VT, Maevskaya MV, Zharkova MS, Kotovskaya YV, Tkacheva ON, Troshina EA, Shestakova MV, Maev IV, Breder VV, Gheivandova NI, Doshchitsin VL, Dudinskaya EN, Ershova EV, Kodzoeva KB, Komshilova KA, Korochanskaya NV, Mayorov AY, Mishina EE, Nadinskaya MY, Nikitin IG, Pogosova NV, Tarzimanova AI, Shamkhalova MS. Clinical Practice Guidelines of the Russian Scientific Liver Society, Russian Gastroenterological Association, Russian Association of Endocrinologists, Russian Association of Gerontologists and Geriatricians and National Society for Preventive Cardiology on Diagnosis and Treatment of Non-Alcoholic Liver Disease. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2022; 32:104-140. [DOI: 10.22416/1382-4376-2022-32-4-104-140] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Aim:present clinical guidelines, aimed at general practitioners, gastroenterologists, cardiologists, endocrinologists, comprise up-to-date methods of diagnosis and treatment of non-alcoholic fatty liver disease.Key points.Nonalcoholic fatty liver disease, the most wide-spread chronic liver disease, is characterized by accumulation of fat by more than 5 % of hepatocytes and presented by two histological forms: steatosis and nonalcoholic steatohepatitis. Clinical guidelines provide current views on pathogenesis of nonalcoholic fatty liver disease as a multisystem disease, methods of invasive and noninvasive diagnosis of steatosis and liver fibrosis, principles of nondrug treatment and pharmacotherapy of nonalcoholic fatty liver disease and associated conditions. Complications of nonalcoholic fatty liver disease include aggravation of cardiometabolic risks, development of hepatocellular cancer, progression of liver fibrosis to cirrhotic stage.Conclusion.Progression of liver disease can be avoided, cardiometabolic risks can be reduced and patients' prognosis — improved by the timely recognition of diagnosis of nonalcoholic fatty liver disease and associated comorbidities and competent multidisciplinary management of these patients.
Collapse
Affiliation(s)
| | | | | | - Yu. V. Kotovskaya
- Russian Gerontology Research and Clinical Centre, Pirogov Russian National Research Medical University
| | - O. N. Tkacheva
- Russian Gerontology Research and Clinical Centre, Pirogov Russian National Research Medical University
| | | | | | - I. V. Maev
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - V. V. Breder
- Blokhin National Medical Research Center of Oncology
| | | | | | - E. N. Dudinskaya
- Russian Gerontology Research and Clinical Centre, Pirogov Russian National Research Medical University
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Troelstra MA, Van Dijk AM, Witjes JJ, Mak AL, Zwirs D, Runge JH, Verheij J, Beuers UH, Nieuwdorp M, Holleboom AG, Nederveen AJ, Gurney-Champion OJ. Self-supervised neural network improves tri-exponential intravoxel incoherent motion model fitting compared to least-squares fitting in non-alcoholic fatty liver disease. Front Physiol 2022; 13:942495. [PMID: 36148303 PMCID: PMC9485997 DOI: 10.3389/fphys.2022.942495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Recent literature suggests that tri-exponential models may provide additional information and fit liver intravoxel incoherent motion (IVIM) data more accurately than conventional bi-exponential models. However, voxel-wise fitting of IVIM results in noisy and unreliable parameter maps. For bi-exponential IVIM, neural networks (NN) were able to produce superior parameter maps than conventional least-squares (LSQ) generated images. Hence, to improve parameter map quality of tri-exponential IVIM, we developed an unsupervised physics-informed deep neural network (IVIM3-NET). We assessed its performance in simulations and in patients with non-alcoholic fatty liver disease (NAFLD) and compared outcomes with bi-exponential LSQ and NN fits and tri-exponential LSQ fits. Scanning was performed using a 3.0T free-breathing multi-slice diffusion-weighted single-shot echo-planar imaging sequence with 18 b-values. Images were analysed for visual quality, comparing the bi- and tri-exponential IVIM models for LSQ fits and NN fits using parameter-map signal-to-noise ratios (SNR) and adjusted R2. IVIM parameters were compared to histological fibrosis, disease activity and steatosis grades. Parameter map quality improved with bi- and tri-exponential NN approaches, with a significant increase in average parameter-map SNR from 3.38 to 5.59 and 2.45 to 4.01 for bi- and tri-exponential LSQ and NN models respectively. In 33 out of 36 patients, the tri-exponential model exhibited higher adjusted R2 values than the bi-exponential model. Correlating IVIM data to liver histology showed that the bi- and tri-exponential NN outperformed both LSQ models for the majority of IVIM parameters (10 out of 15 significant correlations). Overall, our results support the use of a tri-exponential IVIM model in NAFLD. We show that the IVIM3-NET can be used to improve image quality compared to a tri-exponential LSQ fit and provides promising correlations with histopathology similar to the bi-exponential neural network fit, while generating potentially complementary additional parameters.
Collapse
Affiliation(s)
- Marian A. Troelstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Amsterdam, Netherlands
- *Correspondence: Marian A. Troelstra,
| | | | - Julia J. Witjes
- Department of Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Diona Zwirs
- Department of Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Jurgen H. Runge
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam UMC, Amsterdam, Netherlands
| | - Ulrich H. Beuers
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | | | - Aart J. Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | | |
Collapse
|
121
|
Albadawy R, Hasanin AH, Agwa SHA, Hamady S, Aboul-Ela YM, Raafat MH, Kamar SS, Othman M, Yahia YA, Matboli M. Rosavin Ameliorates Hepatic Inflammation and Fibrosis in the NASH Rat Model via Targeting Hepatic Cell Death. Int J Mol Sci 2022; 23:10148. [PMID: 36077546 PMCID: PMC9456245 DOI: 10.3390/ijms231710148] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) represents the most common form of chronic liver disease that urgently needs effective therapy. Rosavin, a major constituent of the Rhodiola Rosea plant of the family Crassulaceae, is believed to exhibit multiple pharmacological effects on diverse diseases. However, its effect on non-alcoholic steatohepatitis (NASH), the progressive form of NAFLD, and the underlying mechanisms are not fully illustrated. AIM Investigate the pharmacological activity and potential mechanism of rosavin treatment on NASH management via targeting hepatic cell death-related (HSPD1/TNF/MMP14/ITGB1) mRNAs and their upstream noncoding RNA regulators (miRNA-6881-5P and lnc-SPARCL1-1:2) in NASH rats. RESULTS High sucrose high fat (HSHF) diet-induced NASH rats were treated with different concentrations of rosavin (10, 20, and 30 mg/kg/day) for the last four weeks of dietary manipulation. The data revealed that rosavin had the ability to modulate the expression of the hepatic cell death-related RNA panel through the upregulation of both (HSPD1/TNF/MMP14/ITGB1) mRNAs and their epigenetic regulators (miRNA-6881-5P and lnc-SPARCL1-1:2). Moreover, rosavin ameliorated the deterioration in both liver functions and lipid profile, and thereby improved the hepatic inflammation, fibrosis, and apoptosis, as evidenced by the decreased protein levels of IL6, TNF-α, and caspase-3 in liver sections of treated animals compared to the untreated NASH rats. CONCLUSION Rosavin has demonstrated a potential ability to attenuate disease progression and inhibit hepatic cell death in the NASH animal model. The produced effect was correlated with upregulation of the hepatic cell death-related (HSPD1, TNF, MMP14, and ITGB1) mRNAs-(miRNA-6881-5P-(lnc-SPARCL1-1:2) RNA panel.
Collapse
Affiliation(s)
- Reda Albadawy
- Department of Gastroenterology, Hepatology & Infectious Disease, Faculty of Medicine, Benha University, Benha 13518, Egypt
| | - Amany Helmy Hasanin
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Sara H. A. Agwa
- Clinical Pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Shaimaa Hamady
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Yasmin M. Aboul-Ela
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Mona Hussien Raafat
- Histology and Cell Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Samaa Samir Kamar
- Histology and Cell Biology Department, Kasralainy Faculty of Medicine, Cairo University, Giza 12613, Egypt
| | - Mohamed Othman
- Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yahia A. Yahia
- Biochemistry Department, Faculty of Pharmacy, Misr University for Science and Technology, Giza 12566, Egypt or
- Chemistry Department, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
| | - Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
122
|
Roeb E, Canbay A, Bantel H, Bojunga J, de Laffolie J, Demir M, Denzer UW, Geier A, Hofmann WP, Hudert C, Karlas T, Krawczyk M, Longerich T, Luedde T, Roden M, Schattenberg J, Sterneck M, Tannapfel A, Lorenz P, Tacke F. [Not Available]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:1346-1421. [PMID: 36100202 DOI: 10.1055/a-1880-2283] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- E Roeb
- Gastroenterologie, Medizinische Klinik II, Universitätsklinikum Gießen und Marburg, Gießen, Deutschland
| | - A Canbay
- Medizinische Klinik, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Deutschland
| | - H Bantel
- Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
| | - J Bojunga
- Medizinische Klinik I Gastroent., Hepat., Pneum., Endokrin., Universitätsklinikum Frankfurt, Frankfurt, Deutschland
| | - J de Laffolie
- Allgemeinpädiatrie und Neonatologie, Zentrum für Kinderheilkunde und Jugendmedizin, Universitätsklinikum Gießen und Marburg, Gießen, Deutschland
| | - M Demir
- Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum und Campus Charité Mitte, Berlin, Deutschland
| | - U W Denzer
- Klinik für Gastroenterologie und Endokrinologie, Universitätsklinikum Gießen und Marburg, Marburg, Deutschland
| | - A Geier
- Medizinische Klinik und Poliklinik II, Schwerpunkt Hepatologie, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - W P Hofmann
- Gastroenterologie am Bayerischen Platz - Medizinisches Versorgungszentrum, Berlin, Deutschland
| | - C Hudert
- Klinik für Pädiatrie m. S. Gastroenterologie, Nephrologie und Stoffwechselmedizin, Charité Campus Virchow-Klinikum - Universitätsmedizin Berlin, Berlin, Deutschland
| | - T Karlas
- Klinik und Poliklinik für Onkologie, Gastroenterologie, Hepatologie, Pneumologie und Infektiologie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - M Krawczyk
- Klinik für Innere Medizin II, Gastroent., Hepat., Endokrin., Diabet., Ern.med., Universitätsklinikum des Saarlandes, Homburg, Deutschland
| | - T Longerich
- Pathologisches Institut, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - T Luedde
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - M Roden
- Klinik für Endokrinologie und Diabetologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - J Schattenberg
- I. Medizinische Klinik und Poliklinik, Universitätsmedizin Mainz, Mainz, Deutschland
| | - M Sterneck
- Klinik für Hepatobiliäre Chirurgie und Transplantationschirurgie, Universitätsklinikum Hamburg, Hamburg, Deutschland
| | - A Tannapfel
- Institut für Pathologie, Ruhr-Universität Bochum, Bochum, Deutschland
| | - P Lorenz
- Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS), Berlin, Deutschland
| | - F Tacke
- Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum und Campus Charité Mitte, Berlin, Deutschland
| |
Collapse
|
123
|
Authors, Collaborators:. Updated S2k Clinical Practice Guideline on Non-alcoholic Fatty Liver Disease (NAFLD) issued by the German Society of Gastroenterology, Digestive and Metabolic Diseases (DGVS) - April 2022 - AWMF Registration No.: 021-025. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:e733-e801. [PMID: 36100201 DOI: 10.1055/a-1880-2388] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
|
124
|
Jia W, Wang K, Zhang S, Lu W, Du A, Li J, Ji L, Xu H. Integrating network pharmacology and in vivo experimental validation to reveal the alleviation of mailuoning oral liquid on non-alcoholic fatty liver disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154326. [PMID: 35853303 DOI: 10.1016/j.phymed.2022.154326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) especially the later stage non-alcoholic steatohepatitis (NASH) seriously endangers human's health and has become a global public health issue in recent years. Mailuoning Oral Liquid (MLN) is a modern traditional Chinese medicine prescription composed by Lonicerae japonicae flos, Achyranthis bidentatae radix, Scrophulariae radix and Dendrobium Caulis. MLN is generally used to treat the syndrome of blood stasis in clinical practice. PURPOSE To observe the alleviation of MLN on NASH in vivo, and explore the possible underlying mechanism. Furthermore, this study also aims to find which Chinese medicinal drug contained in MLN exerts the main pharmacological activity. METHODS NASH model was induced in mice by feeding with methionine and choline deficient (MCD) diet. The effects of MLN on hepatic lipids accumulation, liver inflammation, hepatic fibrosis, and the expression of some molecules were investigated by histological observation, biochemical index analysis, quantitative real-time PCR and western blot. Network pharmacology was applied to predict those involved molecular targets and potential mechanisms, which was further validated in vivo. BODIPY fluorescence staining assay was used to detect cellular lipids accumulation. RESULTS MLN (7.8, 23.4 ml/kg) improved NASH in MCD-fed mice. Network pharmacology results demonstrated that peroxisome proliferator-activated receptor α (PPARα) signaling pathway was crucially involved in the MLN-provided alleviation on NASH. Further experimental validation results showed that MLN increased the expression of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and restored the decreased expression of nuclear PPARα in MCD-fed mice. Further results displayed that Achyranthis bidentatae radix and Lonicerae japonicae flos contributed greatly to the MLN-provided alleviation on NASH in vivo. BODIPY fluorescence staining assay showed that 25R-inokosterone and cynaroside, two compounds from Achyranthis bidentatae radix and Lonicerae japonicae flos, obviously reduced intracellular lipids accumulation in hepatocytes stimulated by non-esterified fatty acid (NEFA). CONCLUSION MLN improved NASH in MCD-fed mice, and the PGC-1α-PPARα signaling pathway was involved in this process. Moreover, Lonicerae japonicae flos and Achyranthis bidentatae radix contained in MLN contributed greatly to the MLN-provided improvement on NASH.
Collapse
Affiliation(s)
- Wangya Jia
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Keke Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shaobo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenxu Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ao Du
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jian Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; Jinling Pharmaceutical Co., Ltd., Nanjing 210009, China
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hong Xu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
125
|
Meijnikman AS, van Olden CC, Aydin Ö, Herrema H, Kaminska D, Lappa D, Männistö V, Tremaroli V, Olofsson LE, de Brauw M, van de Laar A, Verheij J, Gerdes VE, Schwartz TW, Nielsen J, Bäckhed F, Pajukanta P, Pihlajamäki J, Tchkonia T, Kirkland JL, Kuipers F, Nieuwdorp M, Groen AK. Hyperinsulinemia Is Highly Associated With Markers of Hepatocytic Senescence in Two Independent Cohorts. Diabetes 2022; 71:1929-1936. [PMID: 35713877 PMCID: PMC9450852 DOI: 10.2337/db21-1076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/25/2022] [Indexed: 11/13/2022]
Abstract
Cellular senescence is an essentially irreversible growth arrest that occurs in response to various cellular stressors and may contribute to development of type 2 diabetes mellitus and nonalcoholic fatty liver disease (NAFLD). In this article, we investigated whether chronically elevated insulin levels are associated with cellular senescence in the human liver. In 107 individuals undergoing bariatric surgery, hepatic senescence markers were assessed by immunohistochemistry as well as transcriptomics. A subset of 180 participants from the ongoing Finnish Kuopio OBesity Surgery (KOBS) study was used as validation cohort. We found plasma insulin to be highly associated with various markers of cellular senescence in liver tissue. The liver transcriptome of individuals with high insulin revealed significant upregulation of several genes associated with senescence: p21, TGFβ, PI3K, HLA-G, IL8, p38, Ras, and E2F. Insulin associated with hepatic senescence independently of NAFLD and plasma glucose. By using transcriptomic data from the KOBS study, we could validate the association of insulin with p21 in the liver. Our results support a potential role for hyperinsulinemia in induction of cellular senescence in the liver. These findings suggest possible benefits of lowering insulin levels in obese individuals with insulin resistance.
Collapse
Affiliation(s)
- Abraham S. Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Casper C. van Olden
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Ömrüm Aydin
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Hilde Herrema
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Dimitra Lappa
- Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Ville Männistö
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio Finland
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise E. Olofsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maurits de Brauw
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | | | - Joanne Verheij
- Department of Pathology, University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Victor E.A. Gerdes
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Thue W. Schwartz
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Nielsen
- Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Region Västra Götaland, Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio Finland
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | | | - Folkert Kuipers
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Albert K. Groen
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
126
|
Hattori Y, Yamada H, Munetsuna E, Ando Y, Mizuno G, Fujii R, Tsuboi Y, Ichino N, Osakabe K, Sugimoto K, Ishikawa H, Ohashi K, Suzuki K. Increased brain-derived neurotrophic factor in the serum of persons with nonalcoholic fatty liver disease. Endocr J 2022; 69:999-1006. [PMID: 35354697 DOI: 10.1507/endocrj.ej21-0584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The increasing prevalence of nonalcoholic fatty liver disease (NAFLD) is a global health problem. In recent years, the inhibitory effect of brain-derived neurotrophic factor (BDNF) on diabetes mellitus and fatty liver has been clarified. The purpose of this study was to analyze the relationship between serum BDNF and NAFLD which caused by abnormal metabolism of glucose and lipids. This cross-sectional study involved 429 participants (mean age, 63.5 years: men, 38.5%) with low alcohol intake. Of the participants, those who had an increase in echogenicity of the liver parenchyma and hepato-renal contrast on ultrasonography were classified as the NAFLD group (n = 88), and the others were classified as the normal (n = 341) group. The NAFLD group was further classified into a mild group (n = 60) and a severe group (n = 28) based on the intensity of echogenicity and visualization of the hepatic vessels and diaphragm. Median BDNF levels were higher in the NAFLD group than the normal group (35.5 vs. 42.3 ng/mL, p < 0.01). Furthermore, BDNF levels tended to be associated with the severity of NAFLD (p < 0.01). In addition to the univariate analysis, in the sex- and age-adjusted model, there was a significant association between the BDNF levels and NAFLD severity (p < 0.01). The fully adjusted regression analysis also showed a positive association between the serum BDNF level and NAFLD (p < 0.01). These results suggest that NAFLD patients have a compensatory increase in circulating BDNF levels.
Collapse
Affiliation(s)
- Yuji Hattori
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Hiroya Yamada
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Yoshitaka Ando
- Department of Clinical Biochemistry, Fujita Health University School of Medical Science, Toyoake 470-1192, Japan
| | - Genki Mizuno
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University Hospital, Toyoake 470-1192, Japan
| | - Ryosuke Fujii
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Yoshiki Tsuboi
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Naohiro Ichino
- Department of Clinical Physiology and Functional Imaging, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Keisuke Osakabe
- Department of Clinical Physiology and Functional Imaging, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Keiko Sugimoto
- Department of Clinical Physiology and Functional Imaging, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Hiroaki Ishikawa
- Department of Clinical Biochemistry, Fujita Health University School of Medical Science, Toyoake 470-1192, Japan
| | - Koji Ohashi
- Department of Clinical Biochemistry, Fujita Health University School of Medical Science, Toyoake 470-1192, Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| |
Collapse
|
127
|
El Ray A, Paradis V, Montasser A, Elghannam M, Shemis M, Nessim I, Abu-Taleb H, Asselah T, Mohamed A, Poté N, Akl M, Marcellin P. Usefulness of the SAF score to characterize NAFLD/NASH in non-cirrhotic HCV patients. EGYPTIAN LIVER JOURNAL 2022. [DOI: 10.1186/s43066-022-00209-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The SAF score (steatosis, activity, and fibrosis) has been developed for the assessment of the histological severity of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). The aim of this study was to assess the usefulness of the SAF score in a homogenous cohort of Egyptian patients with chronic HCV infection (CHC) without any alcohol consumption and without cirrhosis. We performed a prospective cross-sectional study including 70 consecutive Egyptian patients with chronic HCV infection to assess the usefulness of the SAF score to characterize NAFLD/NASH in non-cirrhotic HCV patients. The inclusion criteria included positive serum anti-HCV IgG antibody and positive HCVRNA, absence of treatment, and absence of cirrhosis (fibrosis score < F4). Patients were divided into two groups: with metabolic syndrome (MS) and without metabolic syndrome (non-MS). All patients were exposed to thorough history taking, full clinical examination, and laboratory and ultrasound assessment. Histopathologic evaluation of the liver biopsy for the assessment of steatosis, activity, grade, and fibrosis stage was assessed by 2 pathologists with experience in liver diseases.
Results
We found that the degree of fibrosis increases with aging. Liver biopsies from CHC patients with metabolic syndrome (MS) exhibited a significantly higher stage of fibrosis than biopsies from those without MS; however, the grade of inflammation did not differ significantly between the two groups. No significant correlation was found between the SAF score and the body mass index (BMI) or serum HCV RNA. No significant relation between SAF score, fibrosis, and MS. No significant relation was found between the MS and the level of HCV viremia.
Conclusion
We concluded that steatosis was associated with the fibrosis stage, independently of MS. This suggests that in this population, steatosis might be more related to HCV infection than to NAFLD and that fibrosis progression might be related, at least in part, to the steatosis process, i.e., virus-associated fatty liver disease (VAFLD).
Collapse
|
128
|
Quantitative Prediction of Steatosis in Patients with Non-Alcoholic Fatty Liver by Means of Hepatic MicroRNAs Present in Serum and Correlating with Hepatic Fat. Int J Mol Sci 2022; 23:ijms23169298. [PMID: 36012565 PMCID: PMC9408888 DOI: 10.3390/ijms23169298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent form of chronic liver disease worldwide, but a reliable non-invasive method to quantify liver steatosis in primary healthcare is not available. Circulating microRNAs have been proposed as biomarkers of severe/advanced NAFLD (steatohepatitis and fibrosis). However, the use of circulating miRNAs to quantitatively assess the % of liver fat in suspected NAFLD patients has not been investigated. We performed global miRNA sequencing in two sets of samples: human livers from organ donors (n = 20), and human sera from biopsy-proven NAFLD patients (n = 23), both with a wide range of steatosis quantified in their liver biopsies. Partial least squares (PLS) regression combined with recursive feature elimination (RFE) was used to select miRNAs associated with steatosis. Moreover, regression models with only 2 or 3 miRNAs, with high biological relevance, were built. Comprehensive microRNA sequencing of liver and serum samples resulted in two sets of abundantly expressed miRNAs (418 in liver and 351 in serum). Pearson correlation analyses indicated that 18% of miRNAs in liver and 14.5% in serum were significantly associated with the amount of liver fat. PLS-RFE models demonstrated that 50 was the number of miRNAs providing the lowest error in both liver and serum models predicting steatosis. Comparison of the two miRNA subsets showed 19 coincident miRNAs that were ranked according to biological significance (guide/passenger strand, relative abundance in liver and serum, number of predicted lipid metabolism target genes, correlation significance, etc.). Among them, miR-10a-5p, miR-98-5p, miR-19a-3p, miR-30e-5p, miR-32-5p and miR-145-5p showed the highest biological relevance. PLS regression models with serum levels of 2−3 of these miRNAs predicted the % of liver fat with errors <5%.
Collapse
|
129
|
Meijnikman AS, Lappa D, Herrema H, Aydin O, Krautkramer KA, Tremaroli V, Olofsson LE, Lundqvist A, Bruin S, Acherman Y, Verheij J, Hjorth S, Gerdes VE, Schwartz TW, Groen AK, Bäckhed F, Nielsen J, Nieuwdorp M. A systems biology approach to study non-alcoholic fatty liver (NAFL) in women with obesity. iScience 2022; 25:104828. [PMID: 35992074 PMCID: PMC9382345 DOI: 10.1016/j.isci.2022.104828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 11/03/2021] [Accepted: 07/20/2022] [Indexed: 11/30/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is now the most frequent global chronic liver disease. Individuals with NAFLD exhibited an increased risk of all-cause mortality driven by extrahepatic cancers and liver and cardiovascular disease. Once the disease is established, women have a higher risk of disease progression and worse outcome. It is therefore critical to deepen the current knowledge on the pathophysiology of NAFLD in women. Here, we used a systems biology approach to investigate the contribution of different organs to this disease. We analyzed transcriptomics profiles of liver and adipose tissues, fecal metagenomes, and plasma metabolomes of 55 women with and without NAFLD. We observed differences in metabolites, expression of human genes, and gut microbial features between the groups and revealed that there is substantial crosstalk between these different omics sets. Multi-omics analysis of individuals with NAFLD may provide novel strategies to study the pathophysiology of NAFLD in humans. The gut microbiome is significantly different between women with and without NAFLD The NAFLD plasma metabolome is characterized by lipid and amino acids post prandial Women with NAFLD have distinct transcriptional profiles in liver and adipose tissue Women with NAFL have a different response upon MMT after massive weight loss
Collapse
Affiliation(s)
- Abraham S. Meijnikman
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
- Corresponding author
| | - Dimitra Lappa
- Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Hilde Herrema
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Omrum Aydin
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Kimberly A. Krautkramer
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Louise E. Olofsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Lundqvist
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sjoerd Bruin
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Yair Acherman
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Joanne Verheij
- Department of Pathology, UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Siv Hjorth
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Victor E.A. Gerdes
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | - Thue W. Schwartz
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Albert K. Groen
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Heath and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden
| | - Jens Nielsen
- Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Corresponding author
| | - Max Nieuwdorp
- Departments of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
- Corresponding author
| |
Collapse
|
130
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease in children. Although environmental factors are major contributors to early onset, children have both shared and unique genetic risk alleles as compared with adults with NAFLD. Treatment relies on reducing environmental risk factors, but many children have persistent diseases. No medications are approved specifically for the treatment of NAFLD, but some anti-obesity or diabetes treatments may be beneficial. Pediatric NAFLD increases the risk of diabetes and other cardiovascular risk factors. Long-term prospective studies are needed to determine the long-term risk of hepatic and non-hepatic morbidity and mortality in adulthood.
Collapse
Affiliation(s)
- Stavra A Xanthakos
- Professor of Pediatrics, Division of Gastroenterology Hepatology and Nutrition, Cincinnati Children's, Department of Pediatrics, Director, Nonalcoholic Steatohepatitis Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
131
|
McPherson S, Armstrong MJ, Cobbold JF, Corless L, Anstee QM, Aspinall RJ, Barclay ST, Brennan PN, Cacciottolo TM, Goldin RD, Hallsworth K, Hebditch V, Jack K, Jarvis H, Johnson J, Li W, Mansour D, McCallum M, Mukhopadhya A, Parker R, Ross V, Rowe IA, Srivastava A, Thiagarajan P, Thompson AI, Tomlinson J, Tsochatzis EA, Yeoman A, Alazawi W. Quality standards for the management of non-alcoholic fatty liver disease (NAFLD): consensus recommendations from the British Association for the Study of the Liver and British Society of Gastroenterology NAFLD Special Interest Group. Lancet Gastroenterol Hepatol 2022; 7:755-769. [PMID: 35490698 PMCID: PMC7614852 DOI: 10.1016/s2468-1253(22)00061-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is common, affecting approximately 25% of the general population. The evidence base for the investigation and management of NAFLD is large and growing, but there is currently little practical guidance to support development of services and delivery of care. To address this, we produced a series of evidence-based quality standard recommendations for the management of NAFLD, with the aim of improving patient care. A multidisciplinary group of experts from the British Association for the Study of the Liver and British Society of Gastroenterology NAFLD Special Interest Group produced the recommendations, which cover: management of people with, or at risk of, NAFLD before the gastroenterology or liver clinic; assessment and investigations in secondary care; and management in secondary care. The quality of evidence for each recommendation was evaluated by the Grading of Recommendation Assessment, Development and Evaluation tool. An anonymous modified Delphi voting process was conducted individually by each member of the group to assess the level of agreement with each statement. Statements were included when agreement was 80% or greater. From the final list of statements, a smaller number of auditable key performance indicators were selected to allow services to benchmark their practice. It is hoped that services will review their practice against our recommendations and key performance indicators and institute service development where needed to improve the care of patients with NAFLD.
Collapse
Affiliation(s)
- Stuart McPherson
- Liver Unit, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Matthew J Armstrong
- Liver Unit, Queen Elizabeth University Hospital Birmingham NHS Trust, Birmingham, UK; NIHR Biomedical Research Centre, University of Birmingham, Birmingham, UK
| | - Jeremy F Cobbold
- Oxford Liver Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; UK NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lynsey Corless
- Department of Gastroenterology, Hepatology and Endoscopy, Hull University Teaching Hospitals, Hull, UK
| | - Quentin M Anstee
- Liver Unit, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Stephen T Barclay
- Walton Liver Clinic, Glasgow Royal Infirmary, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Paul N Brennan
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Tessa M Cacciottolo
- Liver Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Wellcome Trust/MRC Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Robert D Goldin
- Division of Digestive Diseases, Imperial College, London, UK
| | - Kate Hallsworth
- Liver Unit, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Kathryn Jack
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Helen Jarvis
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK; The Bellingham Practice, Northumberland, UK
| | - Jill Johnson
- Liver Unit, Queen Elizabeth University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Wenhao Li
- Barts Liver Centre, Queen Mary University London and Barts Health NHS Trust, London, UK
| | - Dina Mansour
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Queen Elizabeth Hospital, Gateshead NHS Foundation Trust, Gateshead, UK
| | - Mary McCallum
- Digestive Disorders Department, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Ashis Mukhopadhya
- Digestive Disorders Department, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Richard Parker
- Leeds Liver Unit, St James's University Hospital Leeds, Leeds, UK
| | - Valerie Ross
- Barts Liver Centre, Queen Mary University London and Barts Health NHS Trust, London, UK
| | - Ian A Rowe
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK
| | - Ankur Srivastava
- North Bristol Liver Unit, Southmead Hospital, North Bristol Trust, Bristol, UK
| | | | - Alexandra I Thompson
- Centre for Liver and Digestive Disorders, The Royal Infirmary, Edinburgh, Edinburgh, UK
| | - Jeremy Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK
| | - Andrew Yeoman
- Gwent Liver Unit, The Grange University Health Board, Anuerin Bevan Health Board, Wales, UK
| | - William Alazawi
- Barts Liver Centre, Queen Mary University London and Barts Health NHS Trust, London, UK
| |
Collapse
|
132
|
Pais R, Aron-Wisnewsky J, Bedossa P, Ponnaiah M, Oppert JM, Siksik JM, Genser L, Charlotte F, Thabut D, Clement K, Ratziu V. Persistence of severe liver fibrosis despite substantial weight loss with bariatric surgery. Hepatology 2022; 76:456-468. [PMID: 35076966 DOI: 10.1002/hep.32358] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/26/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS It remains unclear to what extent and which components of advanced liver disease improve after bariatric surgery. We herein describe the histological outcome in patients with advanced NASH and its relationship with weight loss and metabolic improvement. APPROACH AND RESULTS One hundred ninety-six patients with advanced NASH underwent bariatric surgery, 66 of whom agreed to a follow-up liver biopsy at 6 ± 3 years (36 with advanced fibrosis [AF] and 30 with high activity [HA] grade without AF). Liver biopsies LBs were centrally read and histological response was defined as the disappearance of AF or HA. Bariatric surgery induced major histological improvement: 29% of patients had normal histology at follow-up biopsy; 74% had NASH resolution without fibrosis progression; and 70% had ≥1 stage fibrosis regression. However, AF persisted in 47% of patients despite NASH resolution and some degree of fibrosis reversal, only evidenced by the EPoS seven-tier staging classification. These patients had lower weight loss and reduced hypertension or diabetes remission rates. Older age and sleeve gastrectomy were the only independent predictors for persistent AF after adjustment for duration of follow-up. All HA patients had major histological improvement: 50% normal histology, 80% NASH resolution, and 86% a ≥1 grade steatosis reduction. Patients with normal liver at follow-up had the largest weight loss and metabolic improvement. Independent predictors of normal liver were amount of weight loss, high histological activity, and the absence of AF before surgery. CONCLUSIONS Although bariatric surgery successfully reverses active steatohepatitis, AF can persist for many years and is associated with lesser weight loss and metabolic improvement. Weight loss alone may not be sufficient to reverse AF.
Collapse
Affiliation(s)
- Raluca Pais
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France.,Institute of Cardiometabolism and Nutrition, Paris, France.,Centre de Recherche Saint Antoine, INSERM UMRS_938, Paris, France
| | - Judith Aron-Wisnewsky
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France.,CRNH Ile de France, INSERM, UMRS U1269, Nutrition and Obesities Systemic Approaches (NutriOmics), Paris, France
| | - Pierre Bedossa
- INSERM UMRS 1138 CRC, Paris, France.,LiverPat, Paris, France
| | | | - Jean-Michel Oppert
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France
| | - Jean-Michel Siksik
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Laurent Genser
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France.,CRNH Ile de France, INSERM, UMRS U1269, Nutrition and Obesities Systemic Approaches (NutriOmics), Paris, France
| | - Frederic Charlotte
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France
| | - Dominique Thabut
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France.,Centre de Recherche Saint Antoine, INSERM UMRS_938, Paris, France
| | - Karine Clement
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France.,CRNH Ile de France, INSERM, UMRS U1269, Nutrition and Obesities Systemic Approaches (NutriOmics), Paris, France
| | - Vlad Ratziu
- Assistance Publique Hôpitaux De Paris, Hôpital Pitié-Salpêtrière, Paris, France.,Sorbonne Université, Paris, France.,Institute of Cardiometabolism and Nutrition, Paris, France.,INSERM UMRS 1138 CRC, Paris, France
| |
Collapse
|
133
|
Wu X, Cheung CKY, Ye D, Chakrabarti S, Mahajan H, Yan S, Song E, Yang W, Lee CH, Lam KSL, Wang C, Xu A. Serum Thrombospondin-2 Levels Are Closely Associated With the Severity of Metabolic Syndrome and Metabolic Associated Fatty Liver Disease. J Clin Endocrinol Metab 2022; 107:e3230-e3240. [PMID: 35532410 DOI: 10.1210/clinem/dgac292] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Metabolic associated fatty liver disease (MAFLD) is the hepatic manifestation of obesity-related metabolic syndrome (MetS). Noninvasive biomarkers for monitoring the progression and severity of these metabolic comorbidities are needed. OBJECTIVES To investigate the associations of serum thrombospondin-2 (TSP2) with MetS and MAFLD severity, and the potential diagnostic value of serum TSP2 for identifying at-risk metabolic associated steatohepatitis (MASH). METHODS Blood samples, clinical data, and liver biopsies were collected from consecutively recruited 252 individuals with morbid obesity receiving bariatric surgery. Histopathology samples of liver biopsies were examined in a blinded fashion by 3 independent pathologists. Serum TSP2 levels were measured by enzyme-linked immunosorbent assay. RESULTS Serum TSP2 levels were significantly elevated in MetS (1.58 [1.07-2.20] ng/mL) compared with non-MetS (1.28 [0.84-1.73] ng/mL; P = .006) in obese patients and positively correlated with increasing number of the MetS components, fasting glucose, glycated hemoglobin, fasting insulin, C-peptide, and homeostatic model assessment of insulin resistance after adjustment of conventional confounders. Serum TSP2 levels differentiated MASH (1.74 [1.32-3.09] ng/mL) from the other non-MASH less severe groups: normal liver (1.41 [1.04-1.63] ng/mL), simple steatosis (1.45 [0.89-1.92] ng/mL), and borderline MASH (1.30 [0.99-2.17] ng/mL) (P < .05). Elevated serum TSP2 was positively associated with the severity of hepatic steatosis, inflammation, fibrosis, and abnormal liver function independent of age, sex and adiposity. Furthermore, high serum TSP2 identified at-risk MASH with area under the operating curve of 0.84 (95% CI 0.70-0.98). CONCLUSION Serum TSP2 is closely associated with severity and progression of MetS and MAFLD, and is a promising noninvasive biomarker for differentiating MASH from benign steatosis and identifying at-risk MASH patients among individuals with obesity.
Collapse
Affiliation(s)
- Xuerui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Cynthia Kwan Yui Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, Canada
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, Canada
| | - Hema Mahajan
- Insititue of Clinical Pathology and Medical Research, Pathology West, NSW Health Pathology, Sydney, NSW 2145, Australia
- University of Sydney, New South Wales, Australia
- Western Sydney University, New South Wales, Australia
| | - Sen Yan
- Dr. Everett Chalmers Hospital, New Brunswick, Canada
| | - Erfei Song
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wah Yang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chi Ho Lee
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Karen Siu Ling Lam
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
134
|
Burko P, Juggath N, Iliasov R, Fedorova M, Nazarova N. A case report of multinodular hepatic steatosis mimicking pseudotumors of the liver. SA J Radiol 2022; 26:2410. [PMID: 35812703 PMCID: PMC9257931 DOI: 10.4102/sajr.v26i1.2410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/28/2022] [Indexed: 11/03/2022] Open
Abstract
Fatty liver disease (FLD) is a common, benign pathology often found incidentally. We present a clinical case in which metastatic liver disease was suspected on initial imaging studies. Following further investigations, a diagnosis of ‘non-alcoholic fatty liver disease (NAFLD), multinodular type’ was postulated. Subsequent histology confirmed the presence of liver steatosis. Multinodular type hepatic steatosis is a rare, but clinically important pathology to identify and differentiate from other multifocal lesions of the liver parenchyma.
Collapse
Affiliation(s)
- Pavel Burko
- Department of Diagnostic Radiology, National Medical Research Center of Rehabilitation and Balneology, Moscow, Russian Federation
- Department of Diagnostic Radiology, Clinic for Diagnostics and Management on Izmaylova, Penza, Russian Federation
| | - Nitin Juggath
- Department of Radiation Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ruslan Iliasov
- Department of Surgery, Clinic for Diagnostics and Management on Izmaylova, Penza, Russian Federation
| | - Mariya Fedorova
- Department of Morphology, Faculty of General Medicine, Penza State University, Institute of Medicine, Penza, Russian Federation
| | - Natalia Nazarova
- Department of Radiology, Penza Institute for Further Training of Physicians - Branch Campus of the Russian Medical Academy of Continuous Professional Education, Penza, Russian Federation
- Department of Diagnostic Radiology, Clinical Hospital №6 named after G.A. Zakharyin, Penza, Russian Federation
| |
Collapse
|
135
|
Dufour JF, Anstee QM, Bugianesi E, Harrison S, Loomba R, Paradis V, Tilg H, Wong VWS, Zelber-Sagi S. Current therapies and new developments in NASH. Gut 2022; 71:gutjnl-2021-326874. [PMID: 35710299 PMCID: PMC9484366 DOI: 10.1136/gutjnl-2021-326874] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/26/2022] [Indexed: 12/13/2022]
Abstract
Non-alcoholic steatohepatitis is becoming the most important aetiology for advanced liver disease. There has been important progress in the field in recent years and the complexity of the pathophysiology of NASH is better understood. Multiple non-invasive circulating and imaging biomarkers have been tested. The importance of lifestyle has been recognised and several drugs are being tested in clinical trials. This review addresses the challenges that healthcare professionals face in the management of NASH patients.
Collapse
Affiliation(s)
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Stephen Harrison
- Pinnacle clinical research, San Antonio, Texas, USA
- Visiting Professor of Hepatology, University of Oxford, Oxford, UK
| | - Rohit Loomba
- Division of Gastroenterology and Epidemiology, University of California at San Diego, La Jolla, California, USA
| | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Shira Zelber-Sagi
- School of Public Health, University of Haifa, Haifa, Israel
- Department of Gastroenterology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| |
Collapse
|
136
|
Puengel T, Lefere S, Hundertmark J, Kohlhepp M, Penners C, Van de Velde F, Lapauw B, Hoorens A, Devisscher L, Geerts A, Boehm S, Zhao Q, Krupinski J, Charles ED, Zinker B, Tacke F. Combined Therapy with a CCR2/CCR5 Antagonist and FGF21 Analogue Synergizes in Ameliorating Steatohepatitis and Fibrosis. Int J Mol Sci 2022; 23:ijms23126696. [PMID: 35743140 PMCID: PMC9224277 DOI: 10.3390/ijms23126696] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 02/05/2023] Open
Abstract
(1) Background: With new potential drug targets emerging, combination therapies appear attractive to treat non-alcoholic steatohepatitis (NASH) and fibrosis. Chemokine receptor CCR2/5 antagonists can improve fibrosis by reducing monocyte infiltration and altering hepatic macrophage subsets. Fibroblast growth factor 21 (FGF21) may improve NASH by modulating lipid and glucose metabolism. We compared effects of single drug to combination treatment as therapeutic strategies against NASH. (2) Methods: We analyzed serum samples and liver biopsies from 85 nonalcoholic fatty liver disease (NAFLD) patients. A CCR2/5 inhibitor (BMS-687681-02-020) and a pegylated FGF21 agonist (BMS-986171) were tested in male C57BL/6J mice subjected to dietary models of NASH and fibrosis (choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) up to 12 weeks; short- (2w) or long-term (6w) treatment). (3) Results: In NAFLD patients, chemokine CCL2 and FGF21 serum levels correlated with inflammatory serum markers, only CCL2 was significantly associated with advanced liver fibrosis. In rodent NASH, CCR2/5 inhibition significantly reduced circulating Ly6C+ monocytes and hepatic monocyte-derived macrophages, alongside reduced hepatic inflammation and fibrosis. FGF21 agonism decreased body weight, liver triglycerides and histological NASH activity. Combination treatment reflected aspects of both compounds upon short- and long-term application, thereby amplifying beneficial effects on all aspects of steatohepatitis and fibrosis. (4) Conclusions: CCR2/5 inhibition blocks hepatic infiltration of inflammatory monocytes, FGF21 agonism improves obesity-related metabolic disorders. Combined therapy ameliorates steatohepatitis and fibrosis more potently than single drug treatment in rodent NASH, corroborating the therapeutic potential of combining these two approaches in NASH patients.
Collapse
Affiliation(s)
- Tobias Puengel
- Department of Hepatology & Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353 Berlin, Germany; (J.H.); (M.K.); (F.T.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Department of Medicine III, RWTH-University Hospital Aachen, 52074 Aachen, Germany;
- Correspondence: (T.P.); (S.L.); Tel.: +49-30-450-630-057 (T.P.); +49-30-450-553-022 (S.L.)
| | - Sander Lefere
- Department of Medicine III, RWTH-University Hospital Aachen, 52074 Aachen, Germany;
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, B-9000 Ghent, Belgium;
- Correspondence: (T.P.); (S.L.); Tel.: +49-30-450-630-057 (T.P.); +49-30-450-553-022 (S.L.)
| | - Jana Hundertmark
- Department of Hepatology & Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353 Berlin, Germany; (J.H.); (M.K.); (F.T.)
| | - Marlene Kohlhepp
- Department of Hepatology & Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353 Berlin, Germany; (J.H.); (M.K.); (F.T.)
| | - Christian Penners
- Department of Medicine III, RWTH-University Hospital Aachen, 52074 Aachen, Germany;
| | | | - Bruno Lapauw
- Department of Endocrinology, Ghent University, B-9000 Ghent, Belgium; (F.V.d.V.); (B.L.)
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, B-9000 Ghent, Belgium;
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, B-9000 Ghent, Belgium;
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, B-9000 Ghent, Belgium;
| | - Stephanie Boehm
- Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.B.); (Q.Z.); (J.K.); (E.D.C.); (B.Z.)
| | - Qihong Zhao
- Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.B.); (Q.Z.); (J.K.); (E.D.C.); (B.Z.)
| | - John Krupinski
- Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.B.); (Q.Z.); (J.K.); (E.D.C.); (B.Z.)
| | - Edgar D. Charles
- Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.B.); (Q.Z.); (J.K.); (E.D.C.); (B.Z.)
| | - Bradley Zinker
- Bristol-Myers Squibb, Princeton, NJ 08540, USA; (S.B.); (Q.Z.); (J.K.); (E.D.C.); (B.Z.)
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353 Berlin, Germany; (J.H.); (M.K.); (F.T.)
| |
Collapse
|
137
|
NAFLD: Mechanisms, Treatments, and Biomarkers. Biomolecules 2022; 12:biom12060824. [PMID: 35740949 PMCID: PMC9221336 DOI: 10.3390/biom12060824] [Citation(s) in RCA: 201] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently renamed metabolic-associated fatty liver disease (MAFLD), is one of the most common causes of liver diseases worldwide. NAFLD is growing in parallel with the obesity epidemic. No pharmacological treatment is available to treat NAFLD, specifically. The reason might be that NAFLD is a multi-factorial disease with an incomplete understanding of the mechanisms involved, an absence of accurate and inexpensive imaging tools, and lack of adequate non-invasive biomarkers. NAFLD consists of the accumulation of excess lipids in the liver, causing lipotoxicity that might progress to metabolic-associated steatohepatitis (NASH), liver fibrosis, and hepatocellular carcinoma. The mechanisms for the pathogenesis of NAFLD, current interventions in the management of the disease, and the role of sirtuins as potential targets for treatment are discussed here. In addition, the current diagnostic tools, and the role of non-coding RNAs as emerging diagnostic biomarkers are summarized. The availability of non-invasive biomarkers, and accurate and inexpensive non-invasive diagnosis tools are crucial in the detection of the early signs in the progression of NAFLD. This will expedite clinical trials and the validation of the emerging therapeutic treatments.
Collapse
|
138
|
Tafur Sánchez CN, Durá Gil M, Alemán Domínguez del Río A, Hernández Pérez CM, Mora Cuadrado N, Gómez de la Cuesta S, Primo Martín D, de Luis Román D, Aller de la Fuente R. Utilidad práctica de los índices no invasivos en la esteatosis hepática metabólica. ENDOCRINOL DIAB NUTR 2022. [PMID: 35787355 DOI: 10.1016/j.endinu.2021.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
139
|
Jin H, Riaz Rajoka MS, Xu X, Liao N, Pang B, Yan L, Liu G, Sun H, Jiang C, Shao D, Barba FJ, Shi J. Potentials of orally supplemented selenium-enriched Lacticaseibacillus rhamnosus to mitigate the lead induced liver and intestinal tract injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 302:119062. [PMID: 35231537 DOI: 10.1016/j.envpol.2022.119062] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/12/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Lead is a metal that exists naturally in the Earth's crust and is a ubiquitous environmental contaminant. The alleviation of lead toxicity is important to keep human health under lead exposure. Biosynthesized selenium nanoparticle (SeNPs) and selenium-enriched Lactobacillus rhamnosus SHA113 (Se-LRS) were developed in this study, and their potentials in alleviating lead-induced injury to the liver and intestinal tract were evaluated in mice by oral administration for 4 weeks. As results, oral intake of lead acetate (150 mg/kg body weight per day) caused more than 50 times and 100 times lead accumulation in blood and the liver, respectively. Liver function was seriously damaged by the lead exposure, which is indicated as the significantly increased lipid accumulation in the liver, enhanced markers of liver function injury in serum, and occurrence of oxidative stress in liver tissues. Serious injury in intestinal tract was also found under lead exposure, as shown by the decrease of intestinal microbiota diversity and occurrence of oxidative stress. Except the lead content in blood and the liver were lowered by 52% and 58%, respectively, oral administration of Se-LRS protected all the other lead-induced injury markers to the normal level. By the comparison with the effects of normal L. rhamnosus SHA113 and the SeNPs isolated from Se-LRS, high protective effects of Se-LRS can be explained as the extremely high efficiency to promote lead excretion via feces by forming insoluble mixture. These findings illustrate the developed selenium-enriched L. rhamnosus can efficiently protect the liver and intestinal tract from injury by lead.
Collapse
Affiliation(s)
- Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Muhammad Shahid Riaz Rajoka
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai, 980-8572, Japan
| | - Xiaoguang Xu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Ning Liao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Lu Yan
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Guanwen Liu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Hui Sun
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China; School of Hospitality Management, Guilin Tourism University, 26 Liangfeng Road, Yanshan District, Guilin City, Guangxi Province, 541006, China
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China
| | - Francisco J Barba
- Nutrition and Food Science Area, Preventive Medicine and Public Health, Food Science, Toxicology and Fo-rensic Medicine Department, Universitat de València, Faculty of Pharmacy, Avda, Vicent Andrés Estellés, s/n, Burjassot, 46100, València, Spain
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province, 710072, China.
| |
Collapse
|
140
|
Kim Y, Hwang J, Bae SH, Hong SS, Chang YW, Kim HJ, Kim SH, Jin SY. Clinical Feasibility of Shear Wave Dispersion Slope for Noninvasive Diagnosis of Nonalcoholic Steatohepatitis in Patients With Morbid Obesity: Preliminary Results Using US Shear Wave Elastography. Ultrasound Q 2022; 38:149-154. [PMID: 35250014 DOI: 10.1097/ruq.0000000000000599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT We aimed to investigate the clinical feasibility of shear wave dispersion slope for assessing nonalcoholic steatohepatitis (NASH) in patients with morbid obesity before bariatric surgery.This prospective study collected data from 25 participants who received liver biopsy during bariatric surgery between February 2019 and December 2020. All participants underwent ultrasonography shear wave elastography before surgery and shear wave speed and shear wave dispersion slope were measured. Liver specimens were evaluated by 1 pathologist scored histologically for nonalcoholic fatty liver disease (NAFLD). Ultrasonography measurements were compared according to histopathologic findings. Diagnostic performance in differentiating NASH from NAFLD was evaluated using the area under the receiver operating characteristic curve (AUC). Median shear wave speed (1.48 vs 1.62 m/s, P = 0.014) and dispersion slope (8.40 vs 11.80 [m/s]/kHz, P = 0.004) were higher in NASH group than in NAFLD group. Shear wave dispersion slope tended to increase step by step as the severity of activity grade (P = 0.032) and hepatic fibrosis (P = 0.015) increased. The AUC of shear wave dispersion slope for differentiating NASH from NAFLD (AUC, 0.83; 95% confidence intervals, 0.66-1.00) was higher than that of shear wave speed (AUC, 0.78; 95% CI, 0.60-0.97), although it did not reach statistical significance (P = 0.729). Shear wave dispersion slope could be a feasible tool for assessing NASH in patients with morbid obesity.
Collapse
Affiliation(s)
- Yeonsoo Kim
- Department of Radiology, Soonchunhyang University College of Medicine, Seoul Hospital
| | - Jiyoung Hwang
- Department of Radiology, Soonchunhyang University College of Medicine, Seoul Hospital
| | - Sung Hwan Bae
- Department of Radiology, Soonchunhyang University College of Medicine, Seoul Hospital
| | - Seong Sook Hong
- Department of Radiology, Soonchunhyang University College of Medicine, Seoul Hospital
| | - Yun-Woo Chang
- Department of Radiology, Soonchunhyang University College of Medicine, Seoul Hospital
| | - Hyun-Joo Kim
- Department of Radiology, Soonchunhyang University College of Medicine, Seoul Hospital
| | - Sang Hyun Kim
- Department of Surgery, Soonchunhyang University College of Medicine, Seoul Hospital
| | - So-Young Jin
- Department of Pathology, Soonchunhyang University College of Medicine, Seoul Hospital, Seoul, Republic of Korea
| |
Collapse
|
141
|
Tafur Sánchez CN, Durá Gil M, Alemán Domínguez Del Río A, Hernández Pérez CM, Mora Cuadrado N, de la Cuesta SG, Primo Martín D, de Luis Román D, de la Fuente RA. The practical utility of non-invasive indices in metabolic hepatic steatosis. ENDOCRINOL DIAB NUTR 2022; 69:418-425. [PMID: 35787355 DOI: 10.1016/j.endien.2022.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/10/2021] [Indexed: 06/15/2023]
Abstract
BACKGROUND Metabolic hepatic steatosis (metHS) is the most frequent cause of chronic liver disease in our environment. The "gold standard" for its diagnosis continues to be liver biopsy, but this is an invasive technique, is not risk-free, and has great interobserver variability, so noninvasive diagnostic methods are necessary. OBJECTIVE To determine the diagnostic accuracy of non-invasive methods based on clinical and analytical data compared to liver biopsy, and to analyse their concordance with each other in the overall cohort and in subpopulations at risk of metHS. METHODS Prospective observational study of 245 patients aged 19-80 years diagnosed with metHS by liver biopsy. Steatosis indices were calculated: FLI (Fatty Liver Index), LAP (Liver Accumulation Product), HSI-(Hepatitis Score Index) and fibrosis indices: Non-alcoholic fatty liver disease fibrosis score (NFS), fibrosis-4 index (FIB-4) and Hepamet Fibrosis Score (HFS). RESULTS The non-invasive steatosis indices showed high sensitivity, and those of fibrosis, high specificity. To assess steatosis, FLI was the most sensitive index in all subpopulations (89-97%), except in women. To assess fibrosis, HFS offers maximum sensitivity in diabetics (86.7%) and is the index with the highest negative predictive value overall. The COR curves for non-invasive indices in steatosis and fibrosis compared to liver biopsy showed greater areas under the curve for the fibrosis indices, with NFS and HFS offering greater diagnostic accuracy (area > 0.8, p < 0.05). HFS also offers high diagnostic sensitivity in the diabetic population. CONCLUSIONS Non-invasive indices of steatosis are more sensitive and those of fibrosis more specific than liver biopsy. NFS and HFS offer the highest diagnostic accuracy, with HFS having the highest negative predictive value.
Collapse
Affiliation(s)
| | - Miguel Durá Gil
- Servicio de Aparato Digestivo, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | | | - Natalia Mora Cuadrado
- Servicio de Aparato Digestivo, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | - David Primo Martín
- Servicio de Aparato Digestivo, Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain; Centro de Investigación de Endocrinología y Nutrición, Valladolid, Spain
| | - Daniel de Luis Román
- Servicio de Aparato Digestivo, Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain; Centro de Investigación de Endocrinología y Nutrición, Valladolid, Spain
| | - Rocío Aller de la Fuente
- Servicio de Aparato Digestivo, Hospital Clínico Universitario de Valladolid, Valladolid, Spain; Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain; Centro de Investigación de Endocrinología y Nutrición, Valladolid, Spain; Grupo de Investigación Biomédica en Medicina Crítica (BioCritic), Valladolid, Spain
| |
Collapse
|
142
|
Anstee QM, Castera L, Loomba R. Impact of non-invasive biomarkers on hepatology practice: Past, present and future. J Hepatol 2022; 76:1362-1378. [PMID: 35589256 DOI: 10.1016/j.jhep.2022.03.026] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022]
Abstract
Over the last two decades, there have been tremendous advances in the non-invasive diagnosis and risk stratification of chronic liver diseases (CLDs). Non-invasive approaches are based on the quantification of biomarkers in serum samples or on the measurement of liver stiffness, using either ultrasound- or magnetic resonance-based elastography techniques. The fibrosis-4 index (non-patented) and enhanced liver fibrosis test (patented) are the most widely adopted serum markers, whereas vibration-controlled transient elastography is the most widely adopted elastography technique. In this review, we discuss the role of non-invasive tests in the current era, as well as their accuracy and how their use in clinical practice has changed the practice of hepatology, including identification of early cirrhosis in patients with risk factors for CLD, diagnosis of portal hypertension, establishing prognosis in compensated cirrhosis, guiding antiviral treatment, and screening for fibrosis and cirrhosis in primary care.
Collapse
Affiliation(s)
- Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Newcastle NIHR Biomedical Research Centre, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.
| | - Laurent Castera
- Université de Paris, UMR1149 (CRI), Inserm, F-75018 Paris, France; Service d'Hépatologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Beaujon, F-92110 Clichy-la-Garenne, France.
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, University of California at San Diego, La Jolla, CA, United States; Herbert Wertheim School of Public Health, University of California at San Diego, La Jolla, CA, United States.
| |
Collapse
|
143
|
Nakamura A, Yamamoto K, Takeda R, Yamada R, Kubo A, Morikawa K, Ando S, Shimazaki T, Izumi T, Umemura M, Kitagataya T, Shigesawa T, Suzuki K, Kimura M, Nakai M, Sho T, Suda G, Natsuizaka M, Ogawa K, Ohnishi S, Sugiyama T, Takeda H, Sakamoto N. The potential of soluble CD14 in discriminating nonalcoholic steatohepatitis from nonalcoholic fatty liver disease. Hepatol Res 2022; 52:508-521. [PMID: 35129841 DOI: 10.1111/hepr.13757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/19/2022] [Accepted: 02/04/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Although various noninvasive markers and prediction formulas for nonalcoholic steatohepatitis (NASH) have been reported, they are of value only in the diagnosis of the advanced fibrosis stage of NASH. In this study, we evaluated soluble CD14 (sCD14) as a diagnostic marker for discriminating NASH from nonalcoholic fatty liver disease (NAFLD) using an animal model and clinical specimens. METHODS Serum sCD14 levels were measured in samples derived from mice with diet-induced NASH and patients using an enzyme-linked immunosorbent assay. Our cohort enrolled 126 patients with liver needle biopsy-proven NAFLD. RESULTS The intestinal defense mechanism in NASH model mice was altered as a consequence of the unique gut environment. Elevated serum levels of sCD14 were observed in mice with diet-induced NASH, and the condition of the liver was exacerbated as a result of exposure to gut-derived endotoxin. We confirmed that the serum sCD14 levels in NAFL patients significantly differed from those in NASH patients. The area under the curve for distinguishing between NAFL and NASH was 0.891. Moreover, we found that serum sCD14 levels were weakly correlated with the inflammation grade based on the NAFLD activity score (NAS), the grade of fibrosis according to the Brunt fibrosis classification, and a positive correlation with the grade of ballooning based on NAS in patients with NAFLD. CONCLUSION sCD14 could be a useful pathophysiological marker and diagnostic adjunct distinguishing NASH from NAFLD. The use of sCD14 may allow the screening and identification of high-risk groups for NASH development and support early therapeutic interventions.
Collapse
Affiliation(s)
- Akihisa Nakamura
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Koji Yamamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan.,Research Division of Molecular Targeting Therapy and Prevention of GI Cancer, Hokkaido University Hospital, Sapporo, Japan
| | - Rei Takeda
- Department of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Ren Yamada
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Akinori Kubo
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Kenichi Morikawa
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Sayaka Ando
- Department of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Tomoe Shimazaki
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Takaaki Izumi
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Machiko Umemura
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Takashi Kitagataya
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Taku Shigesawa
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Kazuharu Suzuki
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Megumi Kimura
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Masato Nakai
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Takuya Sho
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Mitsuteru Natsuizaka
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Koji Ogawa
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Shunsuke Ohnishi
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| | - Toshiro Sugiyama
- Research Division of Molecular Targeting Therapy and Prevention of GI Cancer, Hokkaido University Hospital, Sapporo, Japan
| | - Hiroshi Takeda
- Department of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
144
|
Zhao X, He Y, Liu J, Zhang Q, Liu L, Qu W, Liu Y, Zeng Z, Zhang H, Jia J, Sun L, Wei L, Zhu Z. Impact of living donor liver with steatosis and idiopathic portal inflammation on clinical outcomes in pediatric liver transplantation: Beijing experience. Hepatobiliary Surg Nutr 2022; 11:340-354. [PMID: 35693402 PMCID: PMC9186188 DOI: 10.21037/hbsn-20-685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/24/2020] [Indexed: 01/24/2025]
Abstract
BACKGROUND To evaluate the impact of steatosis and/or idiopathic portal inflammation (IPI) in living donor livers on recipients' clinical outcomes. METHODS We assessed 305 qualified donor liver samples from June 2013 to December 2018. Donors and recipients' clinical characteristics, including follow-up data were retrieved. The graft and overall survival with/without steatosis or portal inflammation were compared by Kaplan-Meier analysis. RESULTS For living donors, the medium age of was 31.2 (28, 35.8) years old; liver histopathology showed macrovesicular steatosis: 0-5% 264/305 (86.6%) and 5-30% 41/305 (13.4%), IPI: no 220/305 (72.1%) and mild 85/305 (27.9%). For recipients, the medium age was 1.0 (0.6, 1.5) years old; the median pediatric-end-stage-liver-disease score was 16 (5.0, 26.0) and medium follow-up time was 32.8 (24.8, 52.0) months. Biliary atresia (69.5%) was the main indication for liver transplantation (LT). CONCLUSIONS The presence of steatosis and portal inflammation of the donor liver did not impact the clinical outcomes including transaminase or bilirubin normalization, short-/long-term complications and recipients' survival. However, recipients with high pediatric-end-stage-liver-disease score (>16) receiving donor liver with portal inflammation, but not steatosis, had trend negative effect on recipients' survival. In conclusion, donor livers with mild steatosis and portal inflammation were qualified for pediatric living donor LT. However, donor liver with mild portal inflammation would better not be allocated to recipients with high pediatric-end-stage-liver-disease score. This study provided new evidence in pediatric living donor liver allocation.
Collapse
Affiliation(s)
- Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Yafei He
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Jimin Liu
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Qian Zhang
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- Clinical Epidemiology and Evidence Base Medicine Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liwei Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Wei Qu
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Ying Liu
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Zhigui Zeng
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Haiming Zhang
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
| | - Liying Sun
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Lin Wei
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| | - Zhijun Zhu
- Liver Transplant Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing, China
| |
Collapse
|
145
|
Non-alcoholic fatty liver disease in adults 2021: A clinical practice guideline of the Italian Association for the Study of the Liver (AISF), the Italian Society of Diabetology (SID) and the Italian Society of Obesity (SIO). Eat Weight Disord 2022; 27:1603-1619. [PMID: 34914079 PMCID: PMC9123074 DOI: 10.1007/s40519-021-01287-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common and emerging liver disease in adults, paralleling the epidemic of obesity and diabetes and leading to worrisome events (hepatocellular carcinoma and end-stage liver disease). In the past years, mounting evidence added insights about epidemiology, natural history, diagnosis and lifestyle-based or drug treatment of NAFLD. In this rapidly evolving scenario, members of the Associazione Italiana per lo Studio del Fegato, the Società Italiana di Diabetologia and the Società Italiana dell'Obesità reviewed current knowledge on NAFLD. The quality of the published evidence is graded, and practical recommendations are made following the rules and the methodology suggested in Italy by the Centro Nazionale per l'Eccellenza delle cure and Istituto Superiore di Sanità. Whenever possible, recommendations are placed within the context the Italian Healthcare system, with reference to specific experience and local diagnostic and management resources.Level of evidence Level of evidence of recommendations for each PICO question were reported according to available evidence.
Collapse
|
146
|
Leung HHW, Puspanathan P, Chan AWH, Nik Mustapha NR, Wong VWS, Chan WK. Reliability of the nonalcoholic steatohepatitis clinical research network and steatosis activity fibrosis histological scoring systems. J Gastroenterol Hepatol 2022; 37:1131-1138. [PMID: 35362158 DOI: 10.1111/jgh.15843] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/29/2022] [Accepted: 03/20/2022] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND AIM We aimed to determine whether lobular inflammation and ballooning grades in the Non-alcoholic Steatohepatitis Clinical Research Network (NASH CRN) scoring system can be directly translated into the same for the Steatosis Activity Fibrosis scoring system (SAF) and to look at intra-observer and inter-observer agreement for each individual histological component and for diagnosis of non-alcoholic steatohepatitis (NASH) using the two scoring systems. METHODS Four pathologists from two Asian centers scored 20 digitalized slides, twice using the NASH CRN, twice using the SAF. Intra-observer and inter-observer agreement was analyzed using Fleiss' kappa, weighted kappa, or Cohen kappa, where appropriate. RESULTS The intra-observer discrepancy rate when using the NASH CRN compared with the SAF was higher than when using the individual scoring system for lobular inflammation (15% comparing both scoring systems vs 10% and 1.8% for the NASH CRN and the SAF, respectively) and hepatocyte ballooning (33.8% vs 12.5% and 5%, respectively), but not for diagnosis of NASH (6.3% vs 6.3% and 0%, respectively). Intra-observer and inter-observer agreement was substantial to almost perfect, except for inter-observer agreement for lobular inflammation and diagnosis of NASH, which was only fair to moderate in most instances. CONCLUSION These findings do not support the direct inter-translation between the NASH CRN and the SAF. However, the diagnosis of NASH during examinations using the NASH CRN may be comparable with diagnosis of NASH using the SAF, vice versa. The inter-observer agreement for lobular inflammation and NASH diagnosis needs to be improved.
Collapse
Affiliation(s)
- Howard Ho-Wai Leung
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Anthony Wing-Hung Chan
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
147
|
Gillessen A, Angelico F, Chen J, Lu L, Lucena MI, Fu Q, Xie Q, Andrade RJ, Xie W, Xu X, Yu Y, Mao YM, Nan Y. Silymarin for Treating Toxic Liver Disease: International Consensus Recommendations. GASTRO HEP ADVANCES 2022; 1:882-893. [PMID: 39131840 PMCID: PMC11307908 DOI: 10.1016/j.gastha.2022.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/09/2022] [Indexed: 08/13/2024]
Abstract
Chronic liver disease (CLD) is a leading health problem impacting the quality of life globally. China shares a major global burden of CLD-including alcoholic liver disease, nonalcoholic fatty liver disease/metabolic dysfunction-associated fatty liver disease, and drug-induced liver injury, except for chronic viral hepatitis. Several exogenous toxins or endogenous metabolic insults trigger hepatic pathology toward steatosis, inflammation, and fibrosis, which, if left untreated, may culminate in liver cirrhosis. Oxidative stress is a common pathomechanism underlying all phenotypes of toxic liver injury; thus, these may be brought under a unified entity, viz. toxic liver disease (TLD). Therefore, a common strategy to treat TLD is to use antioxidants as hepatoprotective agents. The cornerstone for treating fatty liver disease is lifestyle modification, diet, exercise, and behavioral therapy, along with the limited use of pharmacological agents. Available preclinical and clinical evidence indicates that silymarin is a hepatoprotective agent with established antioxidant, anti-inflammatory, antifibrotic effects. An international expert panel of clinicians was convened to discuss combining alcoholic liver disease, nonalcoholic fatty liver disease/metabolic dysfunction-associated fatty liver disease, drug-induced liver injury, and liver cirrhosis under the single definition of TLD, based on the shared pathologic mechanism of oxidative stress. The panel highlighted the significance of silymarin as an antioxidant treatment for TLD.
Collapse
Affiliation(s)
- Anton Gillessen
- Department of Internal Medicine, Herz-Jesu-Hospital, Muenster, Germany
| | - Francesco Angelico
- Department of Public Health and Infectious Diseases, Sapienza University School of Medicine, Rome, Italy
| | - Jun Chen
- Department of Liver Disease Medical Center/Head of the Fourth Department of Liver Disease, Shenzhen Third People's Hospital, Shenzhen, China
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai, China
| | - Maria Isabel Lucena
- Department of Pharmacology, School of Medicine, University of Málaga, Málaga, Spain
| | - Qingchun Fu
- Department of Liver Disease, Centre of Shanghai Public Health Clinical Centre, Shanghai, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai, China
| | - Raul J. Andrade
- Services of Gastroenterology & Clinical Pharmacology, Málaga Biomedical Research Institute, IBIMA, University Hospital, University of Málaga, Málaga, Spain
| | - Wen Xie
- Liver Disease Centre, Beijing Ditan Hospital Capital Medical University, Beijing, China
| | - Xiaoyuan Xu
- Department of Infectious Diseases, Peking University Health Science Centre, Beijing, China
| | - Yanyan Yu
- Department of Infectious Disease, Peking University First Hospital, Beijing, China
| | - Yi-min Mao
- Department of Gastroenterology, Renji Hospital, Shanghai, China
| | - Yuemin Nan
- Department of Liver Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
148
|
Non-invasive tests accurately stratify patients with NAFLD based on their risk of liver-related events. J Hepatol 2022; 76:1013-1020. [PMID: 35063601 DOI: 10.1016/j.jhep.2021.12.031] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 11/20/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Previous studies on the prognostic significance of non-invasive liver fibrosis tests in non-alcoholic fatty liver disease (NAFLD) lack direct comparison to liver biopsy. We aimed to evaluate the prognostic accuracy of fibrosis-4 (FIB4) and vibration-controlled transient elastography (VCTE), compared to liver biopsy, for the prediction of liver-related events (LREs) in NAFLD. METHODS A total of 1,057 patients with NAFLD and baseline FIB4 and VCTE were included in a multicenter cohort. Of these patients, 594 also had a baseline liver biopsy. The main study outcome during follow-up was occurrence of LREs, a composite endpoint combining cirrhosis complications and/or hepatocellular carcinoma. Discriminative ability was evaluated using Harrell's C-index. RESULTS FIB4 and VCTE showed good accuracy for the prediction of LREs, with Harrell's C-indexes >0.80 (0.817 [0.768-0.866] vs. 0.878 [0.835-0.921], respectively, p = 0.059). In the biopsy subgroup, Harrell's C-indexes of histological fibrosis staging and VCTE were not significantly different (0.932 [0.910-0.955] vs. 0.881 [0.832-0.931], respectively, p = 0.164), while both significantly outperformed FIB4 for the prediction of LREs. FIB4 and VCTE were independent predictors of LREs in the whole study cohort. The stepwise FIB4-VCTE algorithm accurately stratified the risk of LREs: compared to patients with "FIB4 <1.30", those with "FIB4 ≥1.30 then VCTE <8.0 kPa" had similar risk of LREs (adjusted hazard ratio [aHR] 1.3; 95% CI 0.3-6.8), whereas the risk of LREs significantly increased in patients with "FIB4 ≥1.30 then VCTE 8.0-12.0 kPa" (aHR 3.8; 95% CI 1.3-10.9), and even more for those with "FIB4 ≥1.30 then VCTE >12.0 kPa" (aHR 12.4; 95% CI 5.1-30.2). CONCLUSION VCTE and FIB4 accurately stratify patients with NAFLD based on their risk of LREs. These non-invasive tests are alternatives to liver biopsy for the identification of patients in need of specialized management. LAY SUMMARY The amount of fibrosis in the liver is closely associated with the risk of liver-related complications in non-alcoholic fatty liver disease (NAFLD). Liver biopsy currently remains the reference standard for the evaluation of fibrosis, but its application is limited by its invasiveness. Therefore, we evaluated the ability of non-invasive liver fibrosis tests to predict liver-related complications in NAFLD. Our results show that the blood test FIB4 and transient elastography stratify the risk of liver-related complications in NAFLD, and that transient elastography has similar prognostic accuracy as liver biopsy. These results support the use of non-invasive liver fibrosis tests instead of liver biopsy for the management of patients with NAFLD.
Collapse
|
149
|
Zhang S, Wu Z, Shi L, Yan S, Huang Z, Lu B, Wang Z, Ji L. 2,3,5,4'-tetrahydroxy-stilbene-2-O-β-D-glucoside ameliorates NAFLD via attenuating hepatic steatosis through inhibiting mitochondrial dysfunction dependent on SIRT5. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153994. [PMID: 35220131 DOI: 10.1016/j.phymed.2022.153994] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/17/2022] [Accepted: 02/13/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is becoming more and more common in clinic in the world, and the study on its mechanism and treatment strategy has already been a research hotspot. Natural chemical compound 2,3,5,4'-tetrahydroxy-stilbene-2-O-β-d-glucoside (TSG) is isolated from Polygonum multiflorum Thunb. that has already been reported to have the lipid-lowering activity. PURPOSE The purpose of this research was to observe the improvement of TSG on methionine and choline deficient (MCD) diet-induced NAFLD in mice and to further elucidate its engaged mechanism. METHODS NAFLD was induced in mice fed by MCD diet for 6 weeks. The accumulation of lipids in hepatocytes was induced by 0.5 mM non-esterified fatty acid (NEFA). Biochemical parameters in serum or livers from mice were tested. Protein and mRNA expression and stability were measured. Mitochondrial dysfunction was analyzed both in vivo and in vitro. The Label-free quantitative proteomic analysis was used to find potential involved key molecules. RESULTS TSG attenuated hepatic parenchymal cells injury, liver inflammatory responses and hepatic fibrosis, and markedly ameliorated liver steatosis in mice from MCD group. In vitro results indicated that TSG reduced the accumulation of cellular lipids in hepatocytes induced by NEFA. TSG reduced reactive oxygen species (ROS) formation and attenuated mitochondrial dysfunction both in vivo and in vitro. The label-free quantitative proteomic analysis predicted the crucial participation of NAD-dependent protein deacylase sirtuin-5 (SIRT5). Next experimental results further evidenced that TSG enhanced SIRT5 expression in mitochondria both in vitro and in vivo. The TSG-supplied inhibition on ROS formation and mitochondrial dysfunction in hepatocytes was disappeared after the application of SIRT5 siRNA. TSG increased the expression and enzymatic activity of carnitine palmitoyltransferase 1A (CPT1A), but this enhance was diminished in hepatocytes transfected with SIRT5 siRNA. Additionally, the TSG-provided inhibition on cellular lipids accumulation was also disappeared in hepatocytes transfected with SIRT5 siRNA. Further results demonstrated that TSG increased SIRT5 expression by regulating its mRNA stability through enhancing the binding of SIRT5 mRNA with serine/arginine-rich splicing factor 2 (SRSF2), which is an RNA-binding protein (RBP). CONCLUSION TSG attenuated liver steatosis and inhibited NAFLD progression through preventing oxidative stress injury and improving mitochondrial dysfunction, and SIRT5 played a key role in this process.
Collapse
Affiliation(s)
- Shaobo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liang Shi
- Department of Pediatric Endocrinology and Genetic Metabolism, Shanghai Institute for Pediatric Research, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Shihao Yan
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
150
|
Franzè MS, Pollicino T, Raimondo G, Squadrito G. Occult hepatitis B virus infection in hepatitis C virus negative chronic liver diseases. Liver Int 2022; 42:963-972. [PMID: 35246933 PMCID: PMC9310828 DOI: 10.1111/liv.15233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 01/09/2022] [Accepted: 02/16/2022] [Indexed: 01/26/2023]
Abstract
Data concerning the prevalence of hepatitis B virus (HBV) occult infection (OBI) varies greatly in the different studies according to the sensitivity and specificity of the diagnostic approaches and the HBV prevalence in the different populations examined. The clinical implications of OBI are still debated. While the impact of OBI in HBV transmission as well as in HBV reactivation under immunosuppression are well established, the role of OBI in liver disease and hepatocellular carcinoma (HCC) development are still not definitively elucidated. It has been hypothesized that OBI might contribute to worsening the liver disease course when other causes of liver damage co-exist. Furthermore, much evidence suggests a role of OBI in the hepato-carcinogenesis processes through both indirect and direct oncogenic mechanisms that might favour HCC development. Data on the OBI clinical implications mainly come from studies performed in patients with hepatitis C virus (HCV) infection. However, HCV prevalence has dramatically fallen in the past years also because of the advent of specific and highly effective direct acting antivirals, with a consequent abrupt change of the worldwide scenario of chronic liver disease. Information about OBI prevalence and possible clinical impact in non-HCV-related liver disease are fragmentary, and the objective of this review is to critically summarize the available data in this field.
Collapse
Affiliation(s)
- Maria Stella Franzè
- Department of Clinical and Experimental MedicineMessina UniversityMessinaItaly
- Division of Medicine and HepatologyMessina University HospitalMessinaItaly
| | - Teresa Pollicino
- Department of Human PathologyMessina UniversityMessinaItaly
- Division of Advanced Diagnostic LaboratoriesMessina University HospitalMessinaItaly
| | - Giovanni Raimondo
- Department of Clinical and Experimental MedicineMessina UniversityMessinaItaly
- Division of Medicine and HepatologyMessina University HospitalMessinaItaly
| | - Giovanni Squadrito
- Department of Clinical and Experimental MedicineMessina UniversityMessinaItaly
- Division of Internal MedicineMessina University HospitalMessinaItaly
| |
Collapse
|