201
|
Galarregui C, Marin-Alejandre BA, Perez-Diaz-Del-Campo N, Cantero I, Monreal JI, Elorz M, Benito-Boillos A, Herrero JI, Tur JA, Martínez JA, Zulet MA, Abete I. Predictive Value of Serum Ferritin in Combination with Alanine Aminotransferase and Glucose Levels for Noninvasive Assessment of NAFLD: Fatty Liver in Obesity (FLiO) Study. Diagnostics (Basel) 2020; 10:diagnostics10110917. [PMID: 33171699 PMCID: PMC7695258 DOI: 10.3390/diagnostics10110917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/03/2020] [Accepted: 11/07/2020] [Indexed: 12/13/2022] Open
Abstract
The identification of affordable noninvasive biomarkers for the diagnosis and characterization of nonalcoholic fatty liver disease (NAFLD) is a major challenge for the research community. This study aimed to explore the usefulness of ferritin as a proxy biomarker of NAFLD condition, alone or in combination with other routine biochemical parameters. Subjects with overweight/obesity and ultrasound-confirmed liver steatosis (n = 112) from the Fatty Liver in Obesity (FLiO) study were assessed. The hepatic evaluation considered magnetic resonance imaging, ultrasonography, and credited routine blood liver biomarkers. Anthropometry and body composition, dietary intake (by means of a validated 137-item food frequency questionnaire), and specific biochemical markers were also determined. Serum ferritin levels were analyzed using a chemiluminescent microparticle immunoassay kit. Lower serum ferritin concentrations were associated with general better liver health and nutritional status. The evaluation of ferritin as a surrogate of liver damage by means of quantile regression analyses showed a positive association with alanine aminotransferase (ALT) (β = 19.21; p ≤ 0.001), liver fat content (β = 8.70; p = 0.008), and hepatic iron (β = 3.76; p ≤ 0.001), after adjusting for potential confounders. In receiver operating characteristic (ROC) analyses, the panel combination of blood ferritin, glucose, and ALT showed the best prediction for liver fat mass (area under the curve (AUC) 0.82). A combination of ferritin and ALT showed the higher predictive ability for estimating liver iron content (AUC 0.73). This investigation demonstrated the association of serum ferritin with liver health as well as with glucose and lipid metabolism markers in subjects with NAFLD. Current findings led to the identification of ferritin as a potential noninvasive predictive biomarker of NAFLD, whose surrogate value increased when combined with other routine biochemical measurements (glucose/ALT).
Collapse
Affiliation(s)
- Cristina Galarregui
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
| | - Bertha Araceli Marin-Alejandre
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
| | - Nuria Perez-Diaz-Del-Campo
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
| | - Irene Cantero
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
| | - J. Ignacio Monreal
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Clinical Chemistry Department, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Mariana Elorz
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Department of Radiology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alberto Benito-Boillos
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Department of Radiology, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - José Ignacio Herrero
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Liver Unit, Clinica Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Josep A. Tur
- Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands & Balearic Islands Institute for Health Research (IDISBA), 07122 Palma, Spain
| | - J. Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - M. Angeles Zulet
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Correspondence: (M.A.Z.); (I.A.); Tel.: +34-948-42-56-00 (ext. 806317) (M.A.Z.); +34-948-42-56-00 (ext. 806357) (I.A.)
| | - Itziar Abete
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain; (C.G.); (B.A.M.-A.); (N.P.-D.-D.-C.); (I.C.); (J.A.M.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (J.I.M.); (M.E.); (A.B.-B.); (J.I.H.)
- Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Correspondence: (M.A.Z.); (I.A.); Tel.: +34-948-42-56-00 (ext. 806317) (M.A.Z.); +34-948-42-56-00 (ext. 806357) (I.A.)
| |
Collapse
|
202
|
Dajani AI, Popovic B. Essential phospholipids for nonalcoholic fatty liver disease associated with metabolic syndrome: A systematic review and network meta-analysis. World J Clin Cases 2020; 8:5235-5249. [PMID: 33269259 PMCID: PMC7674728 DOI: 10.12998/wjcc.v8.i21.5235] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Essential phospholipids (EPL) are used for the supportive treatment of non-alcoholic fatty liver disease (NAFLD), but data are mostly from small-scale studies.
AIM To evaluate the efficacy of EPL treatment in adult patients with NAFLD and type 2 diabetes and/or obesity.
METHODS The MEDLINE, PubMed, Embase, and Cochrane databases were searched up to March 2019 for clinical trials and comparative observational studies. Eligible studies were those published in English or Chinese that enrolled adult patients (≥ 18 years) with NAFLD and type 2 diabetes mellitus and/or obesity receiving EPL as monotherapy or as add-on therapy to existing therapy, and that included at least one of the efficacy outcomes of interest. A variety of studies were identified; thus, direct, indirect and cohort meta-analyses were performed. Mean difference (MD) and 95% confidence interval (CI) were calculated for continuous variables, and relative risk with 95%CI for disease response and recovery. A random-effects model was used to address between-study heterogeneity.
RESULTS Ten studies met the inclusion criteria (n = 22-324). EPL treatment duration ranged from 4 to 72 wk. In the direct meta-analysis (four randomized controlled trials), compared with antidiabetic therapy alone, EPL plus antidiabetic therapy was associated with a significantly greater reduction in [alanine aminotransferase (ALT); MD: 11.28 U/L (95%CI: -17.33, -5.23), P = 0.0003], triglyceride [MD: -49.33 mg/dL (95%CI: -66.43, -32.23), P < 0.0001] and total cholesterol levels [MD: -29.74 mg/dL (95%CI: -38.02, -21.45), P < 0.0001]. There was also a significant increase in the rate of overall improvement [relative risk 1.50 (95%CI: 1.26-1.79), P < 0.0001], and risk of no disease (P = 0.0091), and a reduction in moderate disease (P = 0.0187); there were no significant differences in severe disease, mild disease, or significant improvement. In the cohort meta-analysis of three non-randomized clinical trials, the MD in ALT levels was -16.71 U/L (95%CI: -24.94, -8.49) and 23% of patients had improved disease. In the cohort meta-analysis of five randomized trials, MD in ALT levels was –28.53 U/L (95%CI: -35.42, -21.65), and 87% (95%CI: 81%, 93%) and 58% (95%CI: 46%, 70%) of patients showed clinical improvement and significant clinical improvement.
CONCLUSION This analysis provides evidence for a benefit of EPL in patients with NAFLD and diabetes and/or obesity. Further large-scale trials are warranted.
Collapse
Affiliation(s)
- Asad Izziddin Dajani
- ADSC, Medcare Hospital and Saudi German Hospital, Sharjah, Al Khan, PO Box 6328, United Arab Emirates
| | - Branko Popovic
- Sanofi-Aventis Deutschland GmbH, CHC Global Medical Affairs, Frankfurt am Main 65926, Germany
| |
Collapse
|
203
|
Kage M, Aishima S, Kusano H, Yano H. Histopathological findings of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Med Ultrason (2001) 2020; 47:549-554. [PMID: 33136250 DOI: 10.1007/s10396-020-01061-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is based on the concept of pathological morphology as well as clinical findings, and is broadly categorized into nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH). The differential diagnosis between NAFL and NASH is important because NASH has the potential to progress to cirrhosis and hepatocellular carcinoma. NAFL is simple hepatic steatosis without hepatocellular injury, while NASH is characterized by macrovesicular steatosis, inflammation, and ballooning hepatocytes with a predominantly centrilobular (zone 3) distribution. Liver biopsy is a useful test for diagnosing NAFLD, but it is invasive. Therefore, various noninvasive methods including diagnostic imaging have been developed in recent years. To verify their usefulness, it is necessary to clarify in detail how the pathological findings are reflected in the image findings as imaging and histopathological findings are closely related. We describe the main histological features of NAFLD, i.e., steatosis, inflammation, ballooning hepatocytes, Mallory-Denk bodies, and fibrosis, as well as the evolutional process to liver cirrhosis.
Collapse
Affiliation(s)
- Masayoshi Kage
- Department of Medical Engineering, Junshin Gakuen University, 1-1-1 Chikushigaoka, Minami-ku, Fukuoka, 815-8510, Japan.
| | - Shinichi Aishima
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Hironori Kusano
- Department of Pathology, School of Medicine, Kurume University, Kurume, Japan
| | - Hirohisa Yano
- Department of Pathology, School of Medicine, Kurume University, Kurume, Japan
| |
Collapse
|
204
|
Gu W, Wen K, Yan C, Li S, Liu T, Xu C, Liu L, Zhao M, Zhang J, Geng T, Gong D. Maintaining intestinal structural integrity is a potential protective mechanism against inflammation in goose fatty liver. Poult Sci 2020; 99:5297-5307. [PMID: 33142445 PMCID: PMC7647926 DOI: 10.1016/j.psj.2020.08.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/21/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Overfeeding causes severe steatosis but not inflammation in goose liver, suggesting existence of protective components. Previous studies have shown that some intestinal microbes and their metabolites damage intestinal structural integrity and function, thus causing inflammation in the development of human and mouse nonalcoholic fatty liver disease. Therefore, this study hypothesizes that intestinal structural integrity of goose is maintained during overfeeding, which may provide goose fatty liver a protective mechanism against inflammation. To test this hypothesis, 48 seventy-day-old healthy Landes male geese were overfed (as overfeeding group) or normally fed (as control group). Blood and intestine (jejunum, ileum, and cecum) samples were harvested on the 12th and 24th d of overfeeding. Data showed that goose fatty liver was successfully induced by 24 d of overfeeding. Hematoxylin-eosin staining analysis indicated that the arrangement of villi and crypts in the intestine was orderly, and the intestinal structure was intact with no pathological symptoms in the 2 groups. Enzyme-linked immunosorbent assay and quantitative PCR analysis indicated no significant differences in the expression of tight junction and inflammation-related genes as well as plasma lipopolysaccharide concentration between the groups. Ileal hypertrophy and cecal atrophy were observed in the overfed vs. control geese, probably because of change of sphingolipid metabolism. Activation of apoptotic pathway may help cecum avoid necrosis-induced inflammation. In conclusion, healthy and intact intestine provides a layer of protection for goose fatty liver against inflammation. Sphingolipid metabolism may be involved in the adaptation of ileum and cecum to overfeeding. The hypertrophy of ileum makes it an important contributor to the development of goose fatty liver. The atrophy and decline in the function of cecum may be caused by apoptosis induced by overfeeding.
Collapse
Affiliation(s)
- Wang Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Kang Wen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Chunchi Yan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Shuo Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Tongjun Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Cheng Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Long Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Minmeng Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Jun Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China.
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province 225009, P. R. China.
| |
Collapse
|
205
|
Arab JP, Dirchwolf M, Álvares-da-Silva MR, Barrera F, Benítez C, Castellanos-Fernandez M, Castro-Narro G, Chavez-Tapia N, Chiodi D, Cotrim H, Cusi K, de Oliveira CPMS, Díaz J, Fassio E, Gerona S, Girala M, Hernandez N, Marciano S, Masson W, Méndez-Sánchez N, Leite N, Lozano A, Padilla M, Panduro A, Paraná R, Parise E, Perez M, Poniachik J, Restrepo JC, Ruf A, Silva M, Tagle M, Tapias M, Torres K, Vilar-Gomez E, Costa Gil JE, Gadano A, Arrese M. Latin American Association for the study of the liver (ALEH) practice guidance for the diagnosis and treatment of non-alcoholic fatty liver disease. Ann Hepatol 2020; 19:674-690. [PMID: 33031970 DOI: 10.1016/j.aohep.2020.09.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) currently represents an epidemic worldwide. NAFLD is the most frequently diagnosed chronic liver disease, affecting 20-30% of the general population. Furthermore, its prevalence is predicted to increase exponentially in the next decades, concomitantly with the global epidemic of obesity, type 2 diabetes mellitus (T2DM), and sedentary lifestyle. NAFLD is a clinical syndrome that encompasses a wide spectrum of associated diseases and hepatic complications such as hepatocellular carcinoma (HCC). Moreover, this disease is believed to become the main indication for liver transplantation in the near future. Since NAFLD management represents a growing challenge for primary care physicians, the Asociación Latinoamericana para el Estudio del Hígado (ALEH) has decided to organize this Practice Guidance for the Diagnosis and Treatment of Non-Alcoholic Fatty Liver Disease, written by Latin-American specialists in different clinical areas, and destined to general practitioners, internal medicine specialists, endocrinologists, diabetologists, gastroenterologists, and hepatologists. The main purpose of this document is to improve patient care and awareness of NAFLD. The information provided in this guidance may also be useful in assisting stakeholders in the decision-making process related to NAFLD. Since new evidence is constantly emerging on different aspects of the disease, updates to this guideline will be required in future.
Collapse
Affiliation(s)
- Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Melisa Dirchwolf
- Unidad de Trasplante Hepático, Servicio de Hepatología, Hospital Privado de Rosario, Rosario, Argentina.
| | - Mário Reis Álvares-da-Silva
- Hepatology Division, Hospital de Clinicas de Porto Alegre, Brazil; School of Medicine, Universidade Federal do Rio Grande do Sul, Brazil; Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - Francisco Barrera
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Carlos Benítez
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | | - Graciela Castro-Narro
- Gastroenterology Department, National Institute of Medical Sciences and Nutrition "Salvador Zubirán", Mexico City, Mexico.
| | | | - Daniela Chiodi
- Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Helma Cotrim
- School of Medicine, Federal University of Bahia, Salvador, Bahia, Brazil.
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA.
| | | | - Javier Díaz
- Departamento del Aparato Digestivo, Hospital Edgardo Rebagliati Martins, EsSalud, Lima, Peru.
| | - Eduardo Fassio
- Sección Hígado, Vías Biliares y Páncreas, Servicio de Gastroenterología, Hospital Nacional Profesor Alejandro Posadas, El Palomar, Buenos Aires, Argentina.
| | - Solange Gerona
- Liver Unit, Hospital de Fuerzas Armadas, Montevideo, Uruguay.
| | | | - Nelia Hernandez
- Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | | | - Walter Masson
- Hospital Italiano de Buenos Aires, Buenos Aires, Argentina.
| | | | - Nathalie Leite
- School of Medicine, Internal Medicine Department and Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Adelina Lozano
- Unidad de Hígado, Servicio de Gastroenterología, Hospital Nacional Arzobispo Loayza, Lima, Peru; Universidad Peruana Cayetano Heredia, Lima, Peru.
| | | | - Arturo Panduro
- Department of Molecular Biology in Medicine, Civil Hospital of Guadalajara, Fray Antonio Alcalde, Guadalajara, Jalisco, Mexico.
| | - Raymundo Paraná
- School of Medicine, Federal University of Bahia, Salvador, Bahia, Brazil.
| | - Edison Parise
- Department of Gastroenterology, Federal University of Sao Paulo, Sao Paulo, Brazil.
| | - Marlene Perez
- Hospital General de la Plaza de la Salud, Santo Domingo, Dominican Republic.
| | - Jaime Poniachik
- Sección de Gastroenterología, Hospital Clínico Universidad de Chile, Santiago, Chile.
| | - Juan Carlos Restrepo
- Hepatobiliary and Liver Transplant Program, Hospital Pablo Tobon Uribe-Universidad de Antioquia, Medellín, Colombia; Grupo Gastrohepatologia, Facultad de Medicina, Universidad of Antioquía UdeA, Medellin, Colombia.
| | - Andrés Ruf
- Unidad de Trasplante Hepático, Servicio de Hepatología, Hospital Privado de Rosario, Rosario, Argentina.
| | - Marcelo Silva
- Hepatology and Liver Transplant Unit, Hospital Universitario Austral, Pilar, Argentina.
| | - Martín Tagle
- Facultad de Medicina Alberto Hurtado, Universidad Peruana Cayetano Heredia, Lima, Peru.
| | - Monica Tapias
- Hospital Universitario Fundación Santa Fe de Bogotá, Bogotá, Colombia.
| | - Kenia Torres
- Hospital General de la Plaza de la Salud, Santo Domingo, Dominican Republic.
| | - Eduardo Vilar-Gomez
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | | - Adrian Gadano
- Liver Unit, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina.
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
206
|
Zhou IY, Tanabe KK, Fuchs BC, Caravan P. Collagen-targeted molecular imaging in diffuse liver diseases. Abdom Radiol (NY) 2020; 45:3545-3556. [PMID: 32737546 DOI: 10.1007/s00261-020-02677-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022]
Abstract
Liver fibrosis is a common pathway shared by all progressive chronic liver diseases (CLD) regardless of the underlying etiologies. With liver biopsy being the gold standard in assessing fibrosis degree, there is a large unmet clinical need to develop non-invasive imaging tools that can directly and repeatedly quantify fibrosis throughout the liver for a more accurate assessment of disease burden, progression, and treatment response. Type I collagen is a particularly attractive target for molecular imaging as its excessive deposition is specific to fibrosis, and it is present in concentrations suitable for many imaging modalities. Novel molecular MRI contrast agents designed to bind with collagen provide direct quantification of collagen deposition, which have been validated across animal species and liver injury models. Collagen-targeted molecular imaging probes hold great promise not only as a tool for initial staging and surveillance of fibrosis progression, but also as a marker of fibrosis regression in drug trials.
Collapse
Affiliation(s)
- Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Harvard Medical School, 149 13th St, Boston, MA, 02129, USA
- Institute for Innovation in Imaging (i3), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kenneth K Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA.
- Harvard Medical School, 149 13th St, Boston, MA, 02129, USA.
- Institute for Innovation in Imaging (i3), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
207
|
Zhou IY, Catalano OA, Caravan P. Advances in functional and molecular MRI technologies in chronic liver diseases. J Hepatol 2020; 73:1241-1254. [PMID: 32585160 PMCID: PMC7572718 DOI: 10.1016/j.jhep.2020.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023]
Abstract
MRI has emerged as the most comprehensive non-invasive diagnostic tool for liver diseases. In recent years, the value of MRI in hepatology has been significantly enhanced by a wide range of contrast agents, both clinically available and under development, that add functional information to anatomically detailed morphological images, or increase the distinction between normal and pathological tissues by targeting molecular and cellular events. Several classes of contrast agents are available for contrast-enhanced hepatic MRI, including i) conventional non-specific extracellular fluid contrast agents for assessing tissue perfusion; ii) hepatobiliary-specific contrast agents that are taken up by functioning hepatocytes and excreted through the biliary system for evaluating hepatobiliary function; iii) superparamagnetic iron oxide particles that accumulate in Kupffer cells; and iv) novel molecular contrast agents that are biochemically targeted to specific molecular/cellular processes for staging liver diseases or detecting treatment responses. The use of different functional and molecular MRI methods enables the non-invasive assessment of disease burden, progression, and treatment response in a variety of liver diseases. A high diagnostic performance can be achieved with MRI by combining imaging biomarkers.
Collapse
Affiliation(s)
- Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Institute for Innovation in Imaging (i(3)), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Onofrio A Catalano
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Division of Abdominal Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Harvard Medical School, Boston, MA, USA; Institute for Innovation in Imaging (i(3)), Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
208
|
De Lorenzo S, Tovoli F, Mazzotta A, Vasuri F, Edeline J, Malvi D, Boudjema K, Renzulli M, Jeddou H, D’Errico A, Turlin B, Cescon M, Uguen T, Granito A, Lièvre A, Brandi G. Non-Alcoholic Steatohepatitis as a Risk Factor for Intrahepatic Cholangiocarcinoma and Its Prognostic Role. Cancers (Basel) 2020; 12:3182. [PMID: 33138044 PMCID: PMC7692633 DOI: 10.3390/cancers12113182] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its most aggressive form, non-alcoholic steatohepatitis (NASH), are causing a rise in the prevalence of hepatocellular carcinoma. Data about NAFLD/NASH and intrahepatic cholangiocarcinoma (iCCA) are few and contradictory, coming from population registries that do not correctly distinguish between NAFLD and NASH. We evaluated the prevalence of NAFLD and NASH in peritumoral tissue of resected iCCA (n = 180) and in needle biopsies of matched liver donors. Data of iCCA patients were subsequently analysed to compare NASH-related iCCA (Group A), iCCA arisen in a healthy liver (Group B) or in patients with classical iCCA risk factors (Group C). NASH was found in 22.5% of 129 iCCA patients without known risk factors and in 6.2% of matched controls (risk ratio 3.625, 95% confidence interval 1.723-7.626, p < 0.001), while NAFLD was equally represented in both groups. The overall survival of NASH-related iCCA was inferior to that of patients with healthy liver (38.5 vs. 48.1 months, p = 0.003) and similar to that of patients with known risk factors (31.9 months, p = 0.948), regardless of liver fibrosis. The multivariable Cox regression confirmed NASH as a prognostic factor (hazard ratio 1.773, 95% confidence interval 1.156-2.718, p = 0.009). We concluded that NASH (but not NAFLD) is a risk factor for iCCA and might affect its prognosis. Dissecting NASH from NAFLD by histology is necessary to correctly assess the actual role of these conditions. Prevention protocols for NASH patients should also consider the risk for iCCA and not only HCC. Mechanistic studies aimed to find a direct pathogenic link between NASH and iCCA could add further relevant information.
Collapse
Affiliation(s)
- Stefania De Lorenzo
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| | - Francesco Tovoli
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| | - Alessandro Mazzotta
- Service de Chirurgie Hépatobiliaire et Digestive, Centre Hospitalier Universitaire Pontchaillou Rennes, CIC-INSERM, Université de Rennes, 35000 Rennes, France; (A.M.); (K.B.); (H.J.)
| | - Francesco Vasuri
- Pathology Unit, S. Orsola-Malpighi Bologna Authority Hospital, 40138 Bologna, Italy; (F.V.); (D.M.); (A.D.)
| | - Julien Edeline
- Department of Medical Oncology, Centre Eugène Marquis, 35000 Rennes, France;
| | - Deborah Malvi
- Pathology Unit, S. Orsola-Malpighi Bologna Authority Hospital, 40138 Bologna, Italy; (F.V.); (D.M.); (A.D.)
| | - Karim Boudjema
- Service de Chirurgie Hépatobiliaire et Digestive, Centre Hospitalier Universitaire Pontchaillou Rennes, CIC-INSERM, Université de Rennes, 35000 Rennes, France; (A.M.); (K.B.); (H.J.)
| | - Matteo Renzulli
- Radiology Unit, S. Orsola-Malpighi Bologna Authority Hospital, 40138 Bologna, Italy;
| | - Heithem Jeddou
- Service de Chirurgie Hépatobiliaire et Digestive, Centre Hospitalier Universitaire Pontchaillou Rennes, CIC-INSERM, Université de Rennes, 35000 Rennes, France; (A.M.); (K.B.); (H.J.)
| | - Antonietta D’Errico
- Pathology Unit, S. Orsola-Malpighi Bologna Authority Hospital, 40138 Bologna, Italy; (F.V.); (D.M.); (A.D.)
| | - Bruno Turlin
- Service de Pathologie-Centre Hospitalier Universitaire Pontchaillou Rennes, INSERM Numecan U1241, Université de Rennes, Centre de Ressources Biologiques-BB-0033-00056, 35000 Rennes, France;
| | - Matteo Cescon
- Surgery Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Thomas Uguen
- Service de Hepatologie, Centre Hospitalier Universitaire Pontchaillou, 35000 Rennes, France;
| | - Alessandro Granito
- Division of Internal Medicine, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| | - Astrid Lièvre
- Department of Gastroenterology, Centre Hospitalier Universitaire Pontchaillou, University of Rennes, Inserm U1242, Rennes, France;
| | - Giovanni Brandi
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| |
Collapse
|
209
|
Zhang L, Zhang Z, Li C, Zhu T, Gao J, Zhou H, Zheng Y, Chang Q, Wang M, Wu J, Ran L, Wu Y, Miao H, Zou X, Liang B. S100A11 Promotes Liver Steatosis via FOXO1-Mediated Autophagy and Lipogenesis. Cell Mol Gastroenterol Hepatol 2020; 11:697-724. [PMID: 33075563 PMCID: PMC7841444 DOI: 10.1016/j.jcmgh.2020.10.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is becoming a severe liver disorder worldwide. Autophagy plays a critical role in liver steatosis. However, the role of autophagy in NAFLD remains exclusive and under debate. In this study, we investigated the role of S100 calcium binding protein A11 (S100A11) in the pathogenesis of hepatic steatosis. METHODS We performed liver proteomics in a well-established tree shrew model of NAFLD. The expression of S100A11 in different models of NAFLD was detected by Western blot and/or quantitative polymerase chain reaction. Liver S100A11 overexpression mice were generated by injecting a recombinant adenovirus gene transfer vector through the tail vein and then induced by a high-fat and high-cholesterol diet. Cell lines with S100a11 stable overexpression were established with a recombinant lentiviral vector. The lipid content was measured with either Bodipy staining, Oil Red O staining, gas chromatography, or a triglyceride kit. The autophagy and lipogenesis were detected in vitro and in vivo by Western blot and quantitative polymerase chain reaction. The functions of Sirtuin 1, histone deacetylase 6 (HDAC6), and FOXO1 were inhibited by specific inhibitors. The interactions between related proteins were analyzed by a co-immunoprecipitation assay and immunofluorescence analysis. RESULTS The expression of S100A11 was up-regulated significantly in a time-dependent manner in the tree shrew model of NAFLD. S100A11 expression was induced consistently in oleic acid-treated liver cells as well as the livers of mice fed a high-fat diet and NAFLD patients. Both in vitro and in vivo overexpression of S100A11 could induce hepatic lipid accumulation. Mechanistically, overexpression of S100A11 activated an autophagy and lipogenesis process through up-regulation and acetylation of the transcriptional factor FOXO1, consequently promoting lipogenesis and lipid accumulation in vitro and in vivo. Inhibition of HDAC6, a deacetylase of FOXO1, showed similar phenotypes to S100A11 overexpression in Hepa 1-6 cells. S100A11 interacted with HDAC6 to inhibit its activity, leading to the release and activation of FOXO1. Under S100A11 overexpression, the inhibition of FOXO1 and autophagy could alleviate the activated autophagy as well as up-regulated lipogenic genes. Both FOXO1 and autophagy inhibition and Dgat2 deletion could reduce liver cell lipid accumulation significantly. CONCLUSIONS A high-fat diet promotes liver S100A11 expression, which may interact with HDAC6 to block its binding to FOXO1, releasing or increasing the acetylation of FOXO1, thus activating autophagy and lipogenesis, and accelerating lipid accumulation and liver steatosis. These findings indicate a completely novel S100A11-HDAC6-FOXO1 axis in the regulation of autophagy and liver steatosis, providing potential possibilities for the treatment of NAFLD.
Collapse
Affiliation(s)
- Linqiang Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhiguo Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Tingting Zhu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jing Gao
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yingzhuan Zheng
- College of Life Sciences, Yunnan Normal University, Kunming, Yunnan, China
| | - Qing Chang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Mingshan Wang
- Howard Hughes Medical Institute, Department of Ecology and Evolutionary Biology, University of California Santa Cruz, Santa Cruz, California
| | - Jieyu Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Liyuan Ran
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China; Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Huilai Miao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Xiaoju Zou
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
210
|
Tête A, Gallais I, Imran M, Legoff L, Martin-Chouly C, Sparfel L, Bescher M, Sergent O, Podechard N, Lagadic-Gossmann D. MEHP/ethanol co-exposure favors the death of steatotic hepatocytes, possibly through CYP4A and ADH involvement. Food Chem Toxicol 2020; 146:111798. [PMID: 33022287 DOI: 10.1016/j.fct.2020.111798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 09/17/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
Liver steatosis has been associated with various etiological factors (obesity, alcohol, environmental contaminants). How those factors work together to induce steatosis progression is still scarcely evaluated. Here, we tested whether phthalates could potentiate death of steatotic hepatocytes when combined with ethanol. Pre-steatotic WIF-B9 hepatocytes were co-exposed to mono (2-ethylhexyl) (MEHP, 500 nM; main metabolite of di (2-ethylhexyl) phthalate or DEHP) and ethanol (5 mM) for 5 days. An increased apoptotic death was detected, involving a DNA damage response. Using 4-Methypyrazole to inhibit ethanol metabolism, and CH-223191 to antagonize the AhR receptor, we found that an AhR-dependent increase in alcohol dehydrogenase (ADH) activity was essential for cell death upon MEHP/ethanol co-exposure. Toxicity was also prevented by HET0016 to inhibit the cytochrome P450 4A (CYP4A). Using the antioxidant thiourea, a role for oxidative stress was uncovered, notably triggering DNA damage. Finally, co-exposing the in vivo steatosis model of high fat diet (HFD)-zebrafish larvae to DEHP (2.56 nM)/ethanol (43 mM), induced the pathological progression of liver steatosis alongside an increased Cyp4t8 (human CYP4A homolog) mRNA expression. Altogether, these results further emphasized the deleterious impact of co-exposures to ethanol/environmental pollutant towards steatosis pathological progression, and unraveled a key role for ADH and CYP4A in such effects.
Collapse
Affiliation(s)
- Arnaud Tête
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Muhammad Imran
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Louis Legoff
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Corinne Martin-Chouly
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Lydie Sparfel
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Maëlle Bescher
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
211
|
Zarei A, Stasi C, Mahmoodi M, Masoumi SJ, Zare M, Jalali M. Effect of soy consumption on liver enzymes, lipid profile, anthropometry indices, and oxidative stress in patients with non-alcoholic fatty liver disease: A systematic review and meta-analysis of clinical trials. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:1245-1250. [PMID: 33149855 PMCID: PMC7585529 DOI: 10.22038/ijbms.2020.46854.10797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/08/2020] [Indexed: 11/06/2022]
Abstract
The present systematic review and meta-analysis was conducted to investigate the effects of soy intake on liver enzymes, lipid profile, anthropometry indices, and oxidative stress in non-alcoholic fatty liver disease (NAFLD). A systematic search was undertaken in PubMed, Embase, Scopus, Web of Science, and Cochrane Library covering up to 10 January 2020. A fixed-effect or random-effects models were applied to pool mean difference (MD) and its 95 % confidence intervals (CI). Four clinical trials comprising 234 participants were included in the meta-analysis. Compared to the controls, alanine aminotransferase (ALT) levels (MD=-7.53, 95% CI=[-11.98, -3.08], P=0.001, I2=0.0 %), body weight (MD=-0.77, 95 % CI=[-1.38, -0.16], P=0.01, I2=36.9%), and the concentration of serum Malondialdehyde (MDA) (MD=-0.75, 95% CI=[-1.29, -0.21], P=0.007, I2=63.6%) were significantly changed following soy intake. Lipid profile was not significantly affected by soy intake. Moreover, no evidence of a significant publication bias was found. The present study suggests lowering effects for soy intake on ALT levels, body weight, and MDA in nonalcoholic liver patients. Therefore, further large-scale and well-designed clinical trials are needed to find conclusive findings.
Collapse
Affiliation(s)
- Aida Zarei
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cristina Stasi
- Interdepartmental Hepatology Center MASVE, Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, Italy
| | - Marzieh Mahmoodi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Jalil Masoumi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Zare
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Jalali
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
212
|
Shin J, Yu JH, Jin YJ, Yim HJ, Jung YK, Yang JM, Song DS, Kim YS, Kim SG, Kim DJ, Suk KT, Yoon EL, Lee SS, Kim CW, Kim HY, Jang JY, Jeong SW. Acute-on-chronic liver failure as a major predictive factor for mortality in patients with variceal bleeding. Clin Mol Hepatol 2020; 26:540-553. [PMID: 32937688 PMCID: PMC7641565 DOI: 10.3350/cmh.2020.0034] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/09/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND/AIMS This study examined the risk factors associated with mortality in cirrhotic patients hospitalized with variceal bleeding, and evaluated the effects of acute-on-chronic liver failure (ACLF) on the prognosis of these patients. METHODS This study was retrospectively conducted on patients registered in the Korean acute-on-chronic liver failure study cohort, and on 474 consecutive cirrhotic patients hospitalized with variceal bleeding from January 2013 to December 2013 at 21 university hospitals. ACLF was defined as described by the European Association for the Study of Liver-Chronic Liver Failure Consortium. RESULTS Among a total of 474 patients, 61 patients were diagnosed with ACLF. The cumulative overall survival (OS) rate was lower in the patients with ACLF than in those without (P<0.001), and patients with higher ACLF grades had a lower OS rate (P<0.001). The chronic liver failure-sequential organ failure assessment (CLIF-SOFA) score was identified as a significant prognostic factor in patients hospitalized with variceal bleeding (hazard ratio [HR], 1.40; 95% confidence interval [CI], 1.30-1.50; P<0.001), even in ACLF patients with variceal bleeding (HR, 1.32; 95% CI, 1.19-1.46, P<0.001). Concerning the prediction of the mortality risk at 28- and 90-day using CLIF-SOFA scores, c-statistics were 0.895 (95% CI, 0.829-0.962) and 0.897 (95% CI, 0.842-0.951), respectively, and the optimal cut-off values were 6.5 and 6.5, respectively. CONCLUSION In cirrhotic patients hospitalized with variceal bleeding, the prognosis was poor when accompanied by ACLF, especially depending upon CLIF-SOFA score. CLIF-SOFA model well predicted the 28-day or 90-day mortality for cirrhotic patients who experienced variceal bleeding.
Collapse
Affiliation(s)
- Jongbeom Shin
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Jung Hwan Yu
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Young-Joo Jin
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Hyung Joon Yim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Young Kul Jung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Korea
| | - Jin Mo Yang
- Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Do Seon Song
- Department of Internal Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young Seok Kim
- Department of Internal Medicine, Soonchunhyang University Hospital Bucheon, Bucheon, Korea
| | - Sang Gyune Kim
- Department of Internal Medicine, Soonchunhyang University Hospital Bucheon, Bucheon, Korea
| | - Dong Joon Kim
- Institute of Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Ki Tae Suk
- Institute of Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Eileen L. Yoon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Sang Soo Lee
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Korea
| | - Chang Wook Kim
- Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee Yeon Kim
- Department of Internal Medicine, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Young Jang
- Institute for Digestive Research and Digestive Disease Center, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital, Seoul, Korea
| | - Soung Won Jeong
- Institute for Digestive Research and Digestive Disease Center, Department of Internal Medicine, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital, Seoul, Korea
| |
Collapse
|
213
|
Angelini G, Castagneto-Gissey L, Casella-Mariolo J, Caristo ME, Russo MF, Lembo E, Verrastro O, Stefanizzi G, Marini PL, Casella G, Bornstein SR, Rubino F, Mingrone G. Duodenal-jejunal bypass improves nonalcoholic fatty liver disease independently of weight loss in rodents with diet-induced obesity. Am J Physiol Gastrointest Liver Physiol 2020; 319:G502-G511. [PMID: 32812775 DOI: 10.1152/ajpgi.00357.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of liver-related mortality. NAFLD is associated with obesity, hepatic fat accumulation, and insulin resistance, all of which contribute to its pathophysiology. Weight-loss is the main therapy for NAFLD, and metabolic surgery is the most effective treatment for morbid obesity and its metabolic comorbidities. Although has been reported that Roux-en-Y gastric bypass can reverse NAFLD, it is unclear whether such effects result from reduced weight, from a lower calorie-intake, or from the direct influence of surgery on mechanisms contributing to NAFLD. We aimed to investigate whether gastrointestinal (GI) bypass surgery could induce direct effects on hepatic fat accumulation and insulin resistance, independently of weight reduction. Twenty Wistar rats on a high-fat diet underwent duodenal-jejunal-bypass (DJB) or sham operation and were pair fed (PF) for 15 wk after surgery to obtain a matched weight. Outcome measures include ectopic fat deposition, expression of genes and proteins involved in fat metabolism, insulin-signaling, and gluconeogenesis in liver and muscle. Despite no differences in body weight and calorie intake, DJB showed lower ectopic fat accumulation, improved peripheral and hepatic insulin sensitivity, and enhanced lipid droplet degradation. In both tissues, DJB increased insulin signaling, whereas hepatic key enzymes involved in gluconeogenesis and de novo lipogenesis were decreased. These findings suggest that DJB can reverse, independently of weight loss, ectopic fat deposition and insulin resistance, two features of NAFLD that share a mutual pathway, in which perilipin-2 (PLIN2) seems to be the main player, supporting further investigation into strategies that target the gut to treat metabolic liver diseases.NEW & NOTEWORTHY Our findings suggest that duodenal-jejunal bypass can reverse, independently of weight loss, ectopic fat deposition and insulin resistance, two features of nonalcoholic fatty liver disease that share a mutual pathway, in which perilipin-2 seems to be the main player. Our study supports further investigation into the role of proximal small intestine exclusion in the pathophysiology of nonalcoholic fatty liver disease to uncover less invasive treatments that mimic the effects of metabolic surgery and aims to prevent and treat metabolic liver disease.
Collapse
Affiliation(s)
- Giulia Angelini
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy
| | | | | | - Maria Emiliana Caristo
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy
| | - Maria Francesca Russo
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy
| | - Erminia Lembo
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy
| | - Ornella Verrastro
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy
| | - Gianluigi Stefanizzi
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy
| | - Pier Luigi Marini
- Department of Surgery, Azienda Ospedaliera S. Camillo Forlanini, Rome, Italy
| | - Giovanni Casella
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Stefan R Bornstein
- Department of Medicine III, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Dresden, Germany.,Diabetes and Nutritional Sciences, King's College London, London, United Kingdom
| | - Francesco Rubino
- Diabetes and Nutritional Sciences, King's College London, London, United Kingdom
| | - Geltrude Mingrone
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del S. Cuore, Rome, Italy.,Diabetes and Nutritional Sciences, King's College London, London, United Kingdom
| |
Collapse
|
214
|
Lassailly G, Caiazzo R, Ntandja-Wandji LC, Gnemmi V, Baud G, Verkindt H, Ningarhari M, Louvet A, Leteurtre E, Raverdy V, Dharancy S, Pattou F, Mathurin P. Bariatric Surgery Provides Long-term Resolution of Nonalcoholic Steatohepatitis and Regression of Fibrosis. Gastroenterology 2020; 159:1290-1301.e5. [PMID: 32553765 DOI: 10.1053/j.gastro.2020.06.006] [Citation(s) in RCA: 368] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/28/2020] [Accepted: 06/07/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Studies are needed to determine the long-term effects of bariatric surgery for patients with nonalcoholic steatohepatitis (NASH). We evaluated sequential liver samples, collected the time of bariatric surgery and 1 and 5 years later, to assess the long-term effects of bariatric surgery in patients with NASH. METHODS We performed a prospective study of 180 severely obese patients with biopsy-proven NASH, defined by the NASH clinical research network histologic scores. The patients underwent bariatric surgery at a single center in France and were followed for 5 years. We obtained liver samples from 125 of 169 patients (76%) having reached 1 year and 64 of 94 patients (68%) having reached 5 years after surgery. The primary endpoint was the resolution of NASH without worsening of fibrosis at 5 years. Secondary end points were improvement in fibrosis (reduction of ≥1 stage) at 5 years and regression of fibrosis and NASH at 1 and 5 years. RESULTS At 5 years after bariatric surgery, NASH was resolved, without worsening fibrosis, in samples from 84% of patients (n = 64; 95% confidence interval, 73.1%-92.2%). Fibrosis decreased, compared with baseline, in samples from 70.2% of patients (95% CI, 56.6%-81.6%). Fibrosis disappeared from samples from 56% of all patients (95% CI, 42.4%-69.3%) and from samples from 45.5% of patients with baseline bridging fibrosis. Persistence of NASH was associated with no decrease in fibrosis and less weight loss (reduction in body mass index of 6.3 ± 4.1 kg/m2 in patients with persistent NASH vs reduction of 13.4 ± 7.4 kg/m2; P = .017 with resolution of NASH). Resolution of NASH was observed at 1 year after bariatric surgery in biopsies from 84% of patients, with no significant recurrence between 1 and 5 years (P = .17). Fibrosis began to decrease by 1 year after surgery and continued to decrease until 5 years (P < .001). CONCLUSIONS In a long-term follow-up of patients with NASH who underwent bariatric surgery, we observed resolution of NASH in liver samples from 84% of patients 5 years later. The reduction of fibrosis is progressive, beginning during the first year and continuing through 5 years.
Collapse
Affiliation(s)
- Guillaume Lassailly
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Robert Caiazzo
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Line-Carolle Ntandja-Wandji
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Viviane Gnemmi
- Service d'anatomopathologie, Centre Hospitalier Universitaire de Lille, Université de Lille, INSERM UMR-S 1172, Lille, France
| | - Gregory Baud
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Helene Verkindt
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Massih Ningarhari
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Alexandre Louvet
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Emmanuelle Leteurtre
- Service d'anatomopathologie, Centre Hospitalier Universitaire de Lille, Université de Lille, INSERM UMR-S 1172, Lille, France
| | - Violeta Raverdy
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Sébastien Dharancy
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France
| | - François Pattou
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France.
| | - Philippe Mathurin
- Service des Maladies de l'Appareil Digestif et de la Nutrition, Centre Hospitalier Universitaire de Lille, Lille, France; LIRIC (Lille Inflammation Research International Center) U995, Université de Lille, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire de Lille, Lille, France.
| |
Collapse
|
215
|
Jiang J, Wang A, Zhang X, Wang Y, Wang Q, Zhai M, Huang Y, Qi R. The isonicotinamide cocrystal promotes inhibitory effects of naringenin on nonalcoholic fatty liver disease in mice. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
216
|
Inhibitory Effects of Thymol Isolated from Curcuma longa L. on Adipogenesis in HepG2 Cells. Processes (Basel) 2020. [DOI: 10.3390/pr8091191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a disease associated with metabolic syndromes such as diabetes and obesity, regardless of alcohol consumption, and refers to the accumulation of triacylglycerols in the liver. Thymol (THY) is a vegetable essential oil that is naturally contained in the Zingiberaceae and Lamiaceae families. THY was isolated from Curcuma longa L. The rhizomes of Curcuma longa L. were dried, sliced and extracted with 50% ethanol and then isolated through repeated column chromatography. This study was conducted to investigate the inhibitory effect of THY, even in non-alcoholic fatty liver disease, in relation to the inhibiting hyperlipidemia effect of THY, which was demonstrated in previous studies. Hepatocytes were treated with oleate (OA) containing THY to observe lipid accumulation by Oil Red O staining (ORO). We also tested the effect of THY on triacylglycerols (TG) and total cholesterol (TC) in HepG2 cells. Western blot and real-time RT-PCR using sterol regulatory element-binding protein-1c (SREBP-1c), fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), CCAAT-enhancer-binding protein (C/EBP), proliferator-activated receptor γ (PPARγ), and adenosine monophosphate (AMP)-activated protein kinase (AMPK) expressions were carried out. Consequently, inhibition of lipogenesis by THY (100 μM or 200 μM) in NAFLD treated with OA in HepG2 cells was confirmed. The results of TG and TC experiments confirmed a decrease in the degree of fat accumulation in the liver. Furthermore, inhibition of the SREBP-1c, FAS, ACC, C/EBP and PPARγ expressions that mediated fat accumulation and increased AMPK phosphorylation was observed. Taken together, THY is proposed as a potential natural constituent for the treatment of NAFLD.
Collapse
|
217
|
Yue Y, Liu X, Li Y, Xia B, Yu W. The role of TLR4/MyD88/NF-κB pathway in periodontitis-induced liver inflammation of rats. Oral Dis 2020; 27:1012-1021. [PMID: 32853444 PMCID: PMC8247295 DOI: 10.1111/odi.13616] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/24/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The aim of this study was to clarify the immune mechanism of hepatic injury induced by periodontitis using a rat model. METHODS Twenty-four SPF male Wistar rats were randomly divided into two groups: control group (CG) and periodontitis group (PG). In order to induce experimental periodontitis, we tied the wire ligature around bilateral maxillary first molar of rats. After 8 weeks, the following indicators were valued: gingival index, tooth mobility, probing pocket depth; indexes about oxidative stress and circulating biomarkers; bone retraction by micro-CT analysis; Toll-like receptor 4 (TLR4), myeloid differential protein-88 (MyD88), and nuclear factor kappa B (NF-κB) by qRT-PCR and Western blotting; tumor necrosis factor α (TNF-α), and interleukin-6 (IL-6) by qRT-PCR and immunohistochemical staining; inflammation of periodontal and hepatic tissues by histopathological observation. RESULTS Periodontal indicators and micro-CT results showed the raised levels of inflammatory response and bone retraction in PG compared with CG. The mRNA and protein levels of TLR4, MyD88, NF-κB, TNF-α, and IL-6 have indicated high values in PG versus CG. Histopathological analysis revealed a correlation between periodontitis and hepatic injury. CONCLUSION TLR4/MyD88/NF-κB pathway may play a role in periodontitis-induced liver inflammation of rats.
Collapse
Affiliation(s)
- Yiyun Yue
- Department of Periodontology, Jilin University, Changchun, China
| | - Xinchan Liu
- Department of Geriatric Stomatology, Jilin University, Changchun, China
| | - Yan Li
- Department of Periodontology, Jilin University, Changchun, China
| | - Boyuan Xia
- Department of Periodontology, Jilin University, Changchun, China
| | - Weixian Yu
- Department of Geriatric Stomatology, Jilin University, Changchun, China.,Jilin Provincial Laboratory of Biomedical Engineering, School and Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
218
|
A composite biomarker using multiparametric magnetic resonance imaging and blood analytes accurately identifies patients with non-alcoholic steatohepatitis and significant fibrosis. Sci Rep 2020; 10:15308. [PMID: 32943694 PMCID: PMC7499258 DOI: 10.1038/s41598-020-71995-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is major health burden lacking effective pharmacological therapies. Clinical trials enrol patients with histologically-defined NAFLD (non-alcoholic fatty liver disease) activity score (NAS) ≥ 4 and Kleiner-Brunt fibrosis stage (F) ≥ 2; however, screen failure rates are often high following biopsy. This study evaluated a non-invasive MRI biomarker, iron-corrected T1 mapping (cT1), as a diagnostic pre-screening biomarker for NASH. In a retrospective analysis of 86 biopsy confirmed NAFLD patients we explored the potential of blood and imaging biomarkers, both in isolation and in combination, to discriminate those who have NAS ≥ 4 and F ≥ 2 from those without. Stepwise logistic regression was performed to select the optimal combination of biomarkers, diagnostic accuracy was determined using area under the receiver operator curve and model validated confirmed with and fivefold cross-validation. Results showed that levels of cT1, AST, GGT and fasting glucose were all good predictors of NAS ≥ 4 and F ≥ 2, and the model identified the combination of cT1-AST-fasting glucose (cTAG) as far superior to any individual biomarker (AUC 0.90 [0.84–0.97]). This highlights the potential utility of the composite cTAG score for screening patients prior to biopsy to identify those suitable for NASH clinical trial enrolment.
Collapse
|
219
|
Łukawska A, Kałużny M, Nogalska S, Kubicka E, Kuliczkowska-Płaksej J, Bolanowski M. Polycystic ovary syndrome and non-alcoholic fatty
liver disease: Matched pair or sporadic coexistence? POSTEP HIG MED DOSW 2020. [DOI: 10.5604/01.3001.0014.3920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of liver disease in developed
countries. This condition includes benign non-alcoholic fatty liver disease and non-alcoholic
steatohepatitis with possible fibrosis leading to cirrhosis and hepatocellular carcinoma.
Association of NAFLD and polycystic ovary syndrome (PCOS) has been widely discussed.
Women with PCOS are prone to develop NAFLD more often. PCOS is one of the most common
endocrine disorders among reproductive-age women, characterized by an excess of androgens,
anovulation, and polycystic ovary on ultrasound. Obesity, dyslipidemia and insulin resistance
(IR) are frequently observed in women with PCOS, being also important factors predisposing to
the development of NAFLD. IR may stimulate theca cells to excessive production of androgens,
inhibits the production of sex hormone-binding globulin in the liver, which contributes to the
increase of the bioactive form of testosterone. Hyperandrogenemia also plays an important
role in NAFLD pathogenesis and progression. Androgen excess promotes visceral fat accumulation,
development of dyslipidemia, IR, and contributes to low-grade inflammation. The
pathophysiological associations between PCOS and NAFLD are not fully understood although
it seems reasonable to screen PCOS women for the presence of NAFLD.
Collapse
Affiliation(s)
| | - Marcin Kałużny
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Poland
| | | | - Eliza Kubicka
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Poland
| | | | - Marek Bolanowski
- Department of Endocrinology, Diabetes and Isotope Therapy, Wroclaw Medical University, Poland
| |
Collapse
|
220
|
Kessoku T, Kobayashi T, Ozaki A, Iwaki M, Honda Y, Ogawa Y, Imajo K, Saigusa Y, Yamamoto K, Yamanaka T, Usuda H, Wada K, Yoneda M, Saito S, Nakajima A. Rationale and design of a randomised, double-blind, placebo-controlled, parallel-group, investigator-initiated phase 2a study to investigate the efficacy and safety of elobixibat in combination with cholestyramine for non-alcoholic fatty liver disease. BMJ Open 2020; 10:e037961. [PMID: 32907904 PMCID: PMC7482497 DOI: 10.1136/bmjopen-2020-037961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) pathogenesis involves abnormal metabolism of cholesterol and hepatic accumulation of toxic free-cholesterol. Elobixibat (EXB) inhibits the ileal bile acid (BA) transporter. EXB and cholestyramine (CTM) facilitate the removal of free cholesterol from the liver by decreasing BA recirculation to the liver, thereby stimulating novel BA synthesis from cholesterol. In this randomised, double-blind, placebo-controlled, parallel-group, phase IIa study, we aim to provide a proof-of-concept assessment by evaluating the efficacy and safety of EXB in combination with CTM in patients with NAFLD. METHODS AND ANALYSIS A total of 100 adult patients with NAFLD, diagnosed based on low-density lipoprotein cholesterol (LDL-C) level of >120 mg/dL and liver fat content of ≥8% by MRI-based proton density fat fraction (MRI-PDFF), who meet the inclusion/exclusion criteria will be enrolled. The patients will be randomly assigned to receive the combination therapy of 10 mg EXB and 9 g CTM powder (4 g CTM), 10 mg EXB monotherapy, 9 g CTM powder monotherapy or a placebo treatment (n=25 per group). Blood tests and MRIs will be performed 16 weeks following treatment initiation. The primary study endpoint will be the absolute LDL-C level change at week 16 after treatment initiation. The exploratory endpoint will include absolute changes in the liver fat fraction as measured by MRI-PDFF. This proof-of-concept study will determine whether the combination therapy of EXB and CTM is effective and safe for patients with NAFLD. ETHICS AND DISSEMINATION Ethics approval was obtained from the Ethics Committee of Yokohama City University Hospital before participant enrolment. The results of this study will be submitted for publication in international peer-reviewed journals and the key findings will be presented at international scientific conferences. TRIAL REGISTRATION NUMBER NCT04235205.
Collapse
Affiliation(s)
- Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Palliative Medicine, Yokohama City University Hospital, Yokohama, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Anna Ozaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yasushi Honda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Palliative Medicine, Yokohama City University Hospital, Yokohama, Japan
| | - Yuji Ogawa
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kento Imajo
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Koji Yamamoto
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Haruki Usuda
- Department of Pharmacology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Shimane, Japan
| | - Koichiro Wada
- Department of Pharmacology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Shimane, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoru Saito
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
221
|
Thorp A, Stine JG. Exercise as Medicine: The Impact of Exercise Training on Nonalcoholic Fatty Liver Disease. ACTA ACUST UNITED AC 2020; 19:402-411. [PMID: 33767944 DOI: 10.1007/s11901-020-00543-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Purpose of review Nonalcoholic fatty liver disease (NAFLD) is a leading cause of global liver disease. Because current pharmacologic treatments are ineffective, lifestyle change centered on exercise remains the most effective NAFLD treatment. The aim of this systematic review is to summarize and evaluate the current evidence supporting the use of exercise training as a medical treatment for adult patients with NAFLD. Recent findings At least 150 minutes each week of moderate intensity exercise of any type can improve NAFLD, both with and without modest weight loss. Exercise training reduces hepatic steatosis and liver inflammation, favorably changes body composition, improves vascular endothelial function, increases cardiorespiratory fitness and can lead to histologic response. To date, exercise-based NAFLD trials are limited by small sample size and significant heterogeneity. Summary While several key questions remain unanswered, exercise training will always be an important part of the medical management of patients with NAFLD.
Collapse
Affiliation(s)
- Audrey Thorp
- Department of Medicine, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA, USA
| | - Jonathan G Stine
- Division of Gastroenterology and Hepatology, Department of Medicine, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA, USA
- Department of Public Health Sciences, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA, USA
- Liver Center, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA, USA
- Cancer Institute, The Pennsylvania State University- Milton S. Hershey Medical Center, Hershey PA, USA
| |
Collapse
|
222
|
Does adipose tissue inflammation drive the development of non-alcoholic fatty liver disease in obesity? Clin Res Hepatol Gastroenterol 2020; 44:394-402. [PMID: 32044284 DOI: 10.1016/j.clinre.2019.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 09/09/2019] [Accepted: 10/04/2019] [Indexed: 02/06/2023]
Abstract
Obesity, an increasingly common problem in modern societies, is associated with acquired metabolic disturbances. In this perspective, the development of insulin resistance is now recognized to be initiated by inflammation of the adipose tissue, but the events that lead to this inflammation are still vague. Furthermore, visceral adipose tissue plays a significant role in obesity pathophysiology and in its clinical effects, such as non-alcoholic fatty liver disease (NAFLD). Among the possible mechanisms linking NAFLD and obesity, we focused on Visfatin/NAMPT, mostly produced by macrophages infiltrated in adipose tissue and a biomarker of the inflammatory cascade affecting hepatic inflammation in NAFLD. We also addressed the signalling pathway triggered by the binding of VEGF-B to its receptor, which mediates lipid fluxes throughout the body, being a promising target to prevent ectopic lipid accumulation. We reviewed the available literature on the topic and we suggest a crosstalk between adipose tissue inflammation and NAFLD in order to provide new insights about the putative mechanisms involved in the development of NAFLD in the obesity context. A better understanding of the pathophysiological processes underlying NAFLD will allow the development of new therapeutic approaches.
Collapse
|
223
|
Methods used in the selection of instruments for outcomes included in core outcome sets have improved since the publication of the COSMIN/COMET guideline. J Clin Epidemiol 2020; 125:64-75. [DOI: 10.1016/j.jclinepi.2020.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/21/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
|
224
|
Zeltser N, Meyer I, Hernandez GV, Trahan MJ, Fanter RK, Abo-Ismail M, Glanz H, Strand CR, Burrin DG, La Frano MR, Manjarín R, Maj M. Neurodegeneration in juvenile Iberian pigs with diet-induced nonalcoholic fatty liver disease. Am J Physiol Endocrinol Metab 2020; 319:E592-E606. [PMID: 32744096 PMCID: PMC7864229 DOI: 10.1152/ajpendo.00120.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The objective of this study was to investigate whether juvenile Iberian pigs with diet-induced nonalcoholic fatty liver disease (NAFLD), cholestasis, and gut dysbiosis would develop histological and metabolic markers of neurodegeneration in the frontal cortex (FC) and whether supplementing probiotics would influence the response to the diet. Twenty-eight juvenile Iberian pigs were fed for 10 wk either a control (CON) or high-fructose high-fat (HFF) diet with or without a commercial probiotic mixture. Compared with CON, HFF-fed pigs had a decreased number of neurons and an increase in reactive astrocytes in FC tissue. There was also a decrease in one-carbon metabolites choline and betaine and a marked accumulation of bile acids, cholesteryl esters, and polyol pathway intermediates in FC of HFF-fed pigs, which were associated with markers of neurodegeneration and accentuated with the severity of NAFLD. Betaine depletion in FC tissue was negatively correlated with choline-derived phospholipids in colon content, whereas primary conjugated bile acids in FC were associated with cholestasis. Plasma kynurenine-to-tryptophan quotient, as a marker of indoleamine 2,3-dioxygenase activity, and intestinal dysbiosis were also correlated with neuronal loss and astrogliosis. Recognition memory test and FC levels of amyloid-β and phosphorylated Tau did not differ between diets, whereas probiotics increased amyloid-β and memory loss in HFF-fed pigs. In conclusion, our results show evidence of neurodegeneration in FC of juvenile Iberian pigs and establish a novel pediatric model to investigate the role of gut-liver-brain axis in diet-induced NAFLD.
Collapse
Affiliation(s)
- Nicole Zeltser
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Isabell Meyer
- Institute of Animal Science, University of Bonn, Bonn, Germany
| | - Gabriella V Hernandez
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Matthew J Trahan
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
| | - Rob K Fanter
- College of Agriculture, Food, and Environmental Sciences, California Polytechnic State University, San Luis Obispo, California
- Center for Health Research, California Polytechnic State University, San Luis Obispo, California
| | - Mohammed Abo-Ismail
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Hunter Glanz
- Department of Statistics, California Polytechnic State University, San Luis Obispo, California
| | - Christine R Strand
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
| | - Douglas G Burrin
- United States Department of Agriculture-Agricultural Research Services, Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Michael R La Frano
- Center for Health Research, California Polytechnic State University, San Luis Obispo, California
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, California
| | - Rodrigo Manjarín
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| |
Collapse
|
225
|
Suresh RR, Jain S, Chen Z, Tosh DK, Ma Y, Podszun MC, Rotman Y, Salvemini D, Jacobson KA. Design and in vivo activity of A 3 adenosine receptor agonist prodrugs. Purinergic Signal 2020; 16:367-377. [PMID: 32720036 PMCID: PMC7524976 DOI: 10.1007/s11302-020-09715-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Prodrugs (MRS7422, MRS7476) of highly selective A3 adenosine receptor (AR) agonists Cl-IB-MECA and MRS5698, respectively, were synthesized by succinylation of the 2' and 3' hydroxyl groups, and the parent, active drug was shown to be readily liberated upon incubation with liver esterases. The prodrug MRS7476 had greatly increased aqueous solubility compared with parent MRS5698 and was fully efficacious and with a longer duration than MRS7422 in reversing mouse neuropathic pain (chronic constriction injury model, 3 μmol/kg, p.o.), a known A3AR effect. MRS7476 (5 mg/kg, p.o., twice daily) was found to protect against non-alcoholic steatohepatitis (NASH) in the STAM mouse model, indicated by the NAFLD activity score. Hepatocyte ballooning, IL-10 production, and liver histology were significantly normalized in the MRS7476-treated mice, but not liver fibrosis (no change in ACTA2 levels) or inflammation. Hepatic expression of ADORA3 in human NAFLD patients was 1.9-fold lower compared to normal controls. Adora3 expression determined by qPCR in primary mouse liver was associated with the stellate cells, and its mouse full body A3AR knockout worsened liver markers of inflammation and steatosis. Thus, we have introduced a reversible prodrug strategy that enables water solubility and in vivo activity of masked A3AR agonists in models of two disease conditions.
Collapse
Affiliation(s)
- R. Rama Suresh
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, 9000 Rockville Pike, Bethesda, MD 20892-0810 USA
| | - Shanu Jain
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, 9000 Rockville Pike, Bethesda, MD 20892-0810 USA
| | - Zhoumou Chen
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO USA
| | - Dilip K. Tosh
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, 9000 Rockville Pike, Bethesda, MD 20892-0810 USA
| | - Yanling Ma
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD USA
| | - Maren C. Podszun
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD USA
| | - Yaron Rotman
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO USA
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO USA
| | - Kenneth A. Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, 9000 Rockville Pike, Bethesda, MD 20892-0810 USA
| |
Collapse
|
226
|
Dietary intake of specific amino acids and liver status in subjects with nonalcoholic fatty liver disease: fatty liver in obesity (FLiO) study. Eur J Nutr 2020; 60:1769-1780. [PMID: 32857176 DOI: 10.1007/s00394-020-02370-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE Identification of dietary factors involved in the development and progression of nonalcoholic fatty liver disease (NAFLD) is relevant to the current epidemics of the disease. Dietary amino acids appear to play a key role in the onset and progression of NAFLD. The aim of this study was to analyze potential associations between specific dietary amino acids and variables related to glucose metabolism and hepatic status in adults with overweight/obesity and NAFLD. METHODS One hundred and twelve individuals from the Fatty Liver in Obesity (FLiO) study were evaluated. Liver assessment was carried out by ultrasonography, magnetic resonance imaging and analysis of biochemical parameters. Dietary amino acid intake (aromatic amino acids (AAA); branched-chain amino acids (BCAA); sulfur amino acids (SAA)) was estimated by means of a validated 137-item food frequency questionnaire. RESULTS Higher consumption of these amino acids was associated with worse hepatic health. Multiple adjusted regression models confirmed that dietary AAA, BCAA and SAA were positively associated with liver fat content. AAA and BCAA were positively associated with liver iron concentration. Regarding ferritin levels, a positive association was found with BCAA. Dietary intake of these amino acids was positively correlated with glucose metabolism (glycated hemoglobin, triglyceride and glucose index) although the significance disappeared when potential confounders were included in the model. CONCLUSION These findings suggest that the consumption of specific dietary amino acids might negatively impact on liver status and, to a lesser extent on glucose metabolism in subjects with overweight/obesity and NAFLD. A control of specific dietary amino acid composition should be considered in the management of NAFLD and associated insulin resistance. NCT03183193; June 2017.
Collapse
|
227
|
Baba HA, Theurer S, Canbay A, Schwertheim S, Lainka E, Kälsch J, Wohlschläger J. [Liver transplantation. Current aspects of pretransplantation diagnosis and rejection]. DER PATHOLOGE 2020; 41:505-514. [PMID: 32776226 DOI: 10.1007/s00292-020-00813-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Liver transplantation is an established treatment option for patients with end-stage liver disease. The therapy management of these patients is interdisciplinary and requires pathologists to have both clinical and immunological knowledge. Continuous advances in treatment and increasing clinical experience are accompanied by the further development of pathological transplant diagnostics. This article presents and discusses the latest classification of T‑cell-mediated rejection (TCMR), antibody-mediated rejection (AMR), and aspects of pretransplant diagnostics.
Collapse
Affiliation(s)
- Hideo A Baba
- Institut für Pathologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| | - Sarah Theurer
- Institut für Pathologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland
| | - Ali Canbay
- Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Deutschland
| | - Suzan Schwertheim
- Institut für Pathologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland
| | - Elke Lainka
- Klinik für Kinderheilkunde, Universitätsklinikum Essen, Essen, Deutschland
| | - Julia Kälsch
- Klinik für Gastroenterologie, Universitätsklinikum Essen, Essen, Deutschland
| | - Jeremias Wohlschläger
- Institut für Pathologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland
- MVZ für Pathologie, Diakonissenkrankenhaus Flensburg, Flensburg, Deutschland
| |
Collapse
|
228
|
Ponti F, De Cinque A, Fazio N, Napoli A, Guglielmi G, Bazzocchi A. Ultrasound imaging, a stethoscope for body composition assessment. Quant Imaging Med Surg 2020; 10:1699-1722. [PMID: 32742962 DOI: 10.21037/qims-19-1048] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dysregulation of the human's energy balance, mediated by non-performing endocrine organs (liver, skeletal muscle and adipose tissue, above all), can be related to human metabolic disorders characterized by an impaired body composition (BC), such as obesity and sarcopenia. While it is possible to monitor the BC and its variations at different levels, the tissue-organ composition studies have been proven to provide the most clinically applicable information. Ultrasonography (US), a fast, non-invasive, low-cost and widely available technique, holds great potential in the study of BC, as it can directly measure muscles, organs, visceral and subcutaneous fat tissue in different sections of the abdomen and body, overcoming some limits of anthropometric evaluation and other imaging techniques. Purpose of this review article is to explore the technical aspects and the applied methods of US examination to assess the potential clinical role of this technique in the context of BC characterization, investigating four pivotal topics [abdominal fat compartments, subcutaneous adipose tissue (SAT), skeletal muscle, liver].
Collapse
Affiliation(s)
- Federico Ponti
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Antonio De Cinque
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Specialized, Diagnostic, and Experimental Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Nicola Fazio
- Technology Transfer Office, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandro Napoli
- Department of Radiologic, Oncologic and Pathologic Science, La Sapienza University of Rome, Rome, Italy
| | - Giuseppe Guglielmi
- Department of Radiology, University of Foggia, Foggia, Italy.,Department of Radiology, Scientific Institute "Casa Sollievo della Sofferenza" Hospital, Foggia, Italy
| | - Alberto Bazzocchi
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
229
|
Forsgren MF, Nasr P, Karlsson M, Dahlström N, Norén B, Ignatova S, Sinkus R, Cedersund G, Leinhard OD, Ekstedt M, Kechagias S, Lundberg P. Biomarkers of liver fibrosis: prospective comparison of multimodal magnetic resonance, serum algorithms and transient elastography. Scand J Gastroenterol 2020; 55:848-859. [PMID: 32684060 DOI: 10.1080/00365521.2020.1786599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Accurate biomarkers for quantifying liver fibrosis are important for clinical practice and trial end-points. We compared the diagnostic performance of magnetic resonance imaging (MRI), including gadoxetate-enhanced MRI and 31P-MR spectroscopy, with fibrosis stage and serum fibrosis algorithms in a clinical setting. Also, in a subset of patients, MR- and transient elastography (MRE and TE) was evaluated when available. METHODS Patients were recruited prospectively if they were scheduled to undergo liver biopsy on a clinical indication due to elevated liver enzyme levels without decompensated cirrhosis. Within a month of the clinical work-up, an MR-examination and liver needle biopsy were performed on the same day. Based on late-phase gadoxetate-enhanced MRI, a mathematical model calculated hepatobiliary function (relating to OATP1 and MRP2). The hepatocyte gadoxetate uptake rate (KHep) and the normalised liver-to-spleen contrast ratio (LSC_N10) were also calculated. Nine serum fibrosis algorithms were investigated (GUCI, King's Score, APRI, FIB-4, Lok-Index, NIKEI, NASH-CRN regression score, Forns' score, and NAFLD-fibrosis score). RESULTS The diagnostic performance (AUROC) for identification of significant fibrosis (F2-4) was 0.78, 0.80, 0.69, and 0.78 for MRE, TE, LSC_N10, and GUCI, respectively. For the identification of advanced fibrosis (F3-4), the AUROCs were 0.93, 0.84, 0.81, and 0.82 respectively. CONCLUSION MRE and TE were superior for non-invasive identification of significant fibrosis. Serum fibrosis algorithms developed for specific liver diseases are applicable in this cohort of diverse liver diseases aetiologies. Gadoxetate-MRI was sufficiently sensitive to detect the low function losses associated with fibrosis. None was able to efficiently distinguish between stages within the low fibrosis stages.Lay summaryExcessive accumulation of scar tissue, fibrosis, in the liver is an important aspect in chronic liver disease. To replace the invasive needle biopsy, we have explored non-invasive methods to assess liver fibrosis. In our study we found that elastographic methods, which assess the mechanical properties of the liver, are superior in assessing fibrosis in a clinical setting. Of interest from a clinical trial point-of-view, none of the tested methods was sufficiently accurate to distinguish between adjacent moderate fibrosis stages.
Collapse
Affiliation(s)
- Mikael F Forsgren
- Department of Radiation Physics, Department of and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Patrik Nasr
- Department of Gastroenterology and Hepatology, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Markus Karlsson
- Department of Radiation Physics, Department of and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Nils Dahlström
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Radiology, Department of and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Bengt Norén
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Radiology, Department of and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Simone Ignatova
- Department of Clinical Pathology and Clinical Genetics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ralph Sinkus
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom
| | - Gunnar Cedersund
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Olof Dahlqvist Leinhard
- Department of Radiation Physics, Department of and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Department of Gastroenterology and Hepatology, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Stergios Kechagias
- Department of Gastroenterology and Hepatology, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Department of Radiation Physics, Department of and Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| |
Collapse
|
230
|
Long MT, Massaro JM, Hoffmann U, Benjamin EJ, Naimi TS. Alcohol Use Is Associated With Hepatic Steatosis Among Persons With Presumed Nonalcoholic Fatty Liver Disease. Clin Gastroenterol Hepatol 2020; 18:1831-1841.e5. [PMID: 31734449 PMCID: PMC7569606 DOI: 10.1016/j.cgh.2019.11.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/01/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Many individuals presumed to have nonalcoholic fatty liver disease (NAFLD) consume moderate amounts of alcohol. Little is known about patterns of alcohol use in patients with NAFLD or how drinking behaviors affect liver fat. METHODS We conducted a cross-sectional study of 2475 participants of the Framingham Heart Study with hepatic steatosis, as determined by computed tomography. We performed multivariable-adjusted logistic regression models to evaluate the association between alcohol drinking patterns and hepatic steatosis. Models were adjusted for sociodemographic factors, diet, and the components of the metabolic syndrome. We excluded heavy alcohol users, defined as women who consume more than 14 alcohol drinks per week and men who consume more than 21 alcohol drinks per week. RESULTS In our sample (mean age, 49.8 ± 10.2 y; 50.3% women), the prevalence of hepatic steatosis was 17.5%. The total number of alcohol drinks per week and the maximum drinks consumed per drinking day each were associated with hepatic steatosis (adjusted odds ratio [aOR], 1.15; 95% CI, 1.02-1.29 and aOR 1.15; 95% CI, 1.02-1.30). Binge drinking occurred in 25.4% of individuals with presumed NAFLD and was associated with an increased odds of hepatic steatosis (aOR, 1.45; 95% CI, 1.06-1.98) among alcohol users. In a beverage-specific analysis, alcohol use patterns were associated with hepatic steatosis among beer drinkers, but not among wine drinkers. CONCLUSIONS In a cross-sectional study of participants of the Framingham Heart Study with hepatic steatosis, we observed an association between alcohol use and liver fat, even after excluding heavy alcohol users from our analysis. Alcohol use therefore appears to be a risk factor for NAFLD. Prospective studies are needed to validate these findings and determine if alcohol use should be a focus for research, prevention, and treatment of presumed NAFLD.
Collapse
Affiliation(s)
- Michelle T. Long
- Section of Gastroenterology, Boston Medical Center, Boston University School of Medicine
| | - Joseph M. Massaro
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Emelia J. Benjamin
- Division of Cardiology, Boston Medical Center, Boston University School of Medicine,,National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, Massachusetts
| | - Timothy S. Naimi
- Section of General Internal Medicine, Boston Medical Center, Boston University School of Medicine
| |
Collapse
|
231
|
Matiollo C, Rateke ECDM, de Oliveira KG, Turnes BL, da Silva TE, Maccali C, Latini AS, Narciso-Schiavon JL, Schiavon LL. Elevated neopterin levels are associated with acute-on-chronic liver failure and mortality in patients with liver cirrhosis. Dig Liver Dis 2020; 52:753-760. [PMID: 32434738 DOI: 10.1016/j.dld.2020.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/09/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Macrophage activation plays a central role in hepatic and systemic inflammation and is involved in the pathogenesis of acute-on-chronic liver failure (ACLF). AIMS This study aimed to investigate neopterin levels in patients admitted for acute decompensation (AD) of cirrhosis, evaluating its relationship with ACLF and prognosis. METHODS This prospective cohort study included 205 adult subjects hospitalized for AD of cirrhosis. Twenty-one healthy subjects and 89 patients with stable cirrhosis were evaluated as controls. RESULTS Circulating neopterin was higher in AD as compared to stable cirrhosis and healthy controls (p<0.001). ACLF was independently associated with higher neopterin levels (OR 1.015, 95% CI 1.002-1.028, p = 0.025). In the multivariate Cox regression analysis, neopterin levels (HR = 1.002, IC 95% 1.000-1.004, p = 0.041), Child-Pugh class C, and ACLF were predictors of 30-day survival. Among patients with ACLF, the Kaplan-Meier survival probability was 71.4% in those with neopterin levels < 25 nmol/L and 31.0% if neopterin ≥ 25 nmol/L (p<0.001). CONCLUSIONS Higher circulating neopterin was associated with ACLF in patients hospitalized for AD of cirrhosis. Neopterin levels were also independently predictors of high short-term mortality, especially among patients with ACLF, and could represent a useful biomarker of macrophage activation in clinical practice.
Collapse
Affiliation(s)
- Camila Matiollo
- Laboratório de Análises Clínicas, Hospital Universitário, Universidade Federal de Santa Catarina, Brazil
| | | | - Karina Ghisoni de Oliveira
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Universidade Federal de Santa Catarina, Brazil
| | - Bruna Lenfers Turnes
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Universidade Federal de Santa Catarina, Brazil
| | - Telma Erotides da Silva
- Serviço de Gastroenterologia, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Claudia Maccali
- Serviço de Gastroenterologia, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Alexandra Susana Latini
- Laboratório de Bioenergética e Estresse Oxidativo - LABOX, Universidade Federal de Santa Catarina, Brazil
| | - Janaína Luz Narciso-Schiavon
- Serviço de Gastroenterologia, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - L L Schiavon
- Serviço de Gastroenterologia, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
232
|
Lv X, Dong Y, Hu L, Lu F, Zhou C, Qin S. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) for the management of nonalcoholic fatty liver disease (NAFLD): A systematic review. Endocrinol Diabetes Metab 2020; 3:e00163. [PMID: 32704576 PMCID: PMC7375121 DOI: 10.1002/edm2.163] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/19/2020] [Accepted: 05/23/2020] [Indexed: 12/18/2022] Open
Abstract
There are no licensed drugs for nonalcoholic fatty liver disease (NAFLD), and there is a lack of consensus on the best outcome measures for controlled trials. This systematic review aimed to evaluate the efficacy of GLP-1 RAs in the management of NAFLD, the degree of heterogeneity in trial design and the robustness of conclusions drawn from these clinical trials. We searched publication databases and clinical trial registries through 2 November 2019 for clinical trials with NAFLD. We evaluated improvements in histological findings, noninvasive markers of hepatic steatosis, inflammation, and fibrosis, insulin resistance and anthropometric measures. Our final analysis included 24 clinical trials, comprising 6313 participants with a mean duration of 37 weeks. Four clinical trials, including RCT (n = 1), single-arm studies (n = 2) and case series studies (n = 1), used biopsy-confirmed liver histological change as their end-points. The remaining studies (n = 20) used surrogate end-points. GLP-1 RAs were effective for the improvement in hepatic inflammation, hepatic steatosis and fibrosis. More importantly, GLP-1 RAs showed promise in improving the histological features of NASH. In addition, 8 ongoing trials were identified. In this systematic review of published and ongoing clinical trials of the efficacy of GLP-1RAs for NAFLD, we found that GLP-1 RAs are effective for hepatic steatosis and inflammation, with the potential to reverse fibrosis. Further prospective studies of sufficient duration using histological end-points are needed to fully assess the efficacy of GLP-1 RAs in the management of NAFLD.
Collapse
Affiliation(s)
- Xiaodan Lv
- Department of EndocrinologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Yongqiang Dong
- Department of Thyroid SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Lingling Hu
- Department of EndocrinologyNingbo Medical Center Lihuili Eastern HospitalZhejiangChina
| | - Feiyu Lu
- Department of PaediatricsThe First Hospital of Jilin UniversityChangchunChina
| | - Changyu Zhou
- Department of Gastroenterology and HepatologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Shaoyou Qin
- Department of Gastroenterology and HepatologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
233
|
Gesnik M, Bhatt M, Roy Cardinal MH, Destrempes F, Allard L, Nguyen BN, Alquier T, Giroux JF, Tang A, Cloutier G. In vivo Ultrafast Quantitative Ultrasound and Shear Wave Elastography Imaging on Farm-Raised Duck Livers during Force Feeding. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1715-1726. [PMID: 32381381 DOI: 10.1016/j.ultrasmedbio.2020.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 06/11/2023]
Abstract
Shear wave elastography (speed and dispersion), local attenuation coefficient slope and homodyned-K parametric imaging were used for liver steatosis grading. These ultrasound biomarkers rely on physical interactions between shear and compression waves with tissues at both macroscopic and microscopic scales. These techniques were applied in a context not yet studied with ultrasound imaging, that is, monitoring steatosis of force-fed duck livers from pre-force-fed to foie gras stages. Each estimated feature presented a statistically significant trend along the feeding process (p values <10-3). However, whereas a monotonic increase in the shear wave speed was observed along the process, most quantitative ultrasound features exhibited an absolute maximum value halfway through the process. As the liver fat fraction in foie gras is much higher than that seen clinically, we hypothesized that a change in the ultrasound scattering regime is encountered for high-fat fractions, and consequently, care has to be taken when applying ultrasound biomarkers to grading of severe states of steatosis.
Collapse
Affiliation(s)
- Marc Gesnik
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada
| | - Manish Bhatt
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada
| | - Marie-Hélène Roy Cardinal
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada
| | - François Destrempes
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada
| | - Louise Allard
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada
| | - Bich N Nguyen
- Service of Pathology, University of Montreal Hospital (CHUM), Montréal, QC, Canada
| | - Thierry Alquier
- CRCHUM and Montreal Diabetes Research Center, Montréal, QC, Canada; Department of Medicine, University of Montreal, Montréal, QC, Canada
| | - Jean-François Giroux
- Department of Biological Sciences, University of Quebec in Montreal, Montréal, QC, Canada
| | - An Tang
- Service of Radiology, University of Montreal Hospital (CHUM), Montréal, QC, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, University of Montreal, Montréal, QC, Canada; Laboratory of Medical Image Analysis, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, QC, Canada
| | - Guy Cloutier
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center (CRCHUM), Montréal, QC, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, University of Montreal, Montréal, QC, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, QC, Canada.
| |
Collapse
|
234
|
Albhaisi S, Sanyal AJ. Applying Non-Invasive Fibrosis Measurements in NAFLD/NASH: Progress to Date. Pharmaceut Med 2020; 33:451-463. [PMID: 31933238 DOI: 10.1007/s40290-019-00305-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has now become a worldwide health issue due to the obesity epidemic, affecting approximately 90% of the obese population and 15-40% of the general population. It is the most common form of chronic liver disease in the United States. NAFLD constitutes a spectrum of diseases ranging in severity from mild, such as fatty liver, progressing into nonalcoholic steatohepatitis (NASH), then fibrosis, and ending with cirrhosis. NASH and increasing fibrosis stage are associated with increased morbidity and mortality; the fibrosis stage is therefore a critical element of risk stratification needed to determine therapeutic approach and also the response to treatment. Liver biopsy is considered the 'gold standard' in the diagnosis of NAFLD. However, it is not practical for widespread clinical use because it is invasive, costly, and associated with complications including occasional death. These limitations have driven the development of noninvasive tests that can accurately predict the fibrosis stage in those with NAFLD. In this review, we provide a concise overview of different non-invasive measurements used for NAFLD/NASH.
Collapse
Affiliation(s)
- Somaya Albhaisi
- Department of Internal Medicine, Virginia Commonwealth University, Box 980102, Richmond, VA, 23298, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Box 980341, Richmond, VA, 23298, USA.
| |
Collapse
|
235
|
Liu F, Goh GBB, Tiniakos D, Wee A, Leow WQ, Zhao JM, Rao HY, Wang XX, Wang Q, Wan WK, Lim KH, Romero-Gomez M, Petta S, Bugianesi E, Tan CK, Harrison SA, Anstee QM, Chang PEJ, Wei L. qFIBS: An Automated Technique for Quantitative Evaluation of Fibrosis, Inflammation, Ballooning, and Steatosis in Patients With Nonalcoholic Steatohepatitis. Hepatology 2020; 71:1953-1966. [PMID: 31600834 DOI: 10.1002/hep.30986] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 09/24/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Nonalcoholic steatohepatitis (NASH) is a common cause of chronic liver disease. Clinical trials use the NASH Clinical Research Network (CRN) system for semiquantitative histological assessment of disease severity. Interobserver variability may hamper histological assessment, and diagnostic consensus is not always achieved. We evaluate a second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) imaging-based tool to provide an automated quantitative assessment of histological features pertinent to NASH. APPROACH AND RESULTS Images were acquired by SHG/TPEF from 219 nonalcoholic fatty liver disease (NAFLD)/NASH liver biopsy samples from seven centers in Asia and Europe. These were used to develop and validate qFIBS, a computational algorithm that quantifies key histological features of NASH. qFIBS was developed based on in silico analysis of selected signature parameters for four cardinal histopathological features, that is, fibrosis (qFibrosis), inflammation (qInflammation), hepatocyte ballooning (qBallooning), and steatosis (qSteatosis), treating each as a continuous rather than categorical variable. Automated qFIBS analysis outputs showed strong correlation with each respective component of the NASH CRN scoring (P < 0.001; qFibrosis [r = 0.776], qInflammation [r = 0.557], qBallooning [r = 0.533], and qSteatosis [r = 0.802]) and high area under the receiver operating characteristic curve values (qFibrosis [0.870-0.951; 95% confidence interval {CI}, 0.787-1.000; P < 0.001], qInflammation [0.820-0.838; 95% CI, 0.726-0.933; P < 0.001), qBallooning [0.813-0.844; 95% CI, 0.708-0.957; P < 0.001], and qSteatosis [0.939-0.986; 95% CI, 0.867-1.000; P < 0.001]) and was able to distinguish differing grades/stages of histological disease. Performance of qFIBS was best when assessing degree of steatosis and fibrosis, but performed less well when distinguishing severe inflammation and higher ballooning grades. CONCLUSIONS qFIBS is an automated tool that accurately quantifies the critical components of NASH histological assessment. It offers a tool that could potentially aid reproducibility and standardization of liver biopsy assessments required for NASH therapeutic clinical trials.
Collapse
Affiliation(s)
- Feng Liu
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People's Hospital, Beijing, China
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Dina Tiniakos
- Institute of Clinical and Translational Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Pathology, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aileen Wee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, National University Hospital, Singapore
| | - Wei-Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Jing-Min Zhao
- Department of Pathology and Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui-Ying Rao
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People's Hospital, Beijing, China
| | - Xiao-Xiao Wang
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People's Hospital, Beijing, China
| | - Qin Wang
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People's Hospital, Beijing, China
| | - Wei-Keat Wan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Kiat-Hon Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Manuel Romero-Gomez
- Unit for the Clinical Management of Digestive Diseases, Centro para la Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (CIBEREHD), Institute of Biomedicine Seville (IBIS), Virgen del Rocio University Hospital, University of Seville, Seville, Spain
| | - Salvatore Petta
- Sezione di Gastroenterologia ed Epatologia, Dipartimento di Medicina Interna e Specialistica, DIBIMIS, Universita di Palermo, Palermo, Italy
| | - Elisabetta Bugianesi
- Division of Gastroenterology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Chee-Kiat Tan
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Stephen A Harrison
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Quentin M Anstee
- Institute of Clinical and Translational Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Pik-Eu Jason Chang
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Lai Wei
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People's Hospital, Beijing, China.,Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China.,Institute for Precision Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
236
|
Yılmaz Y, Kanı HT, Demirtaş CÖ, Kaya E, Sapmaz AF, Qutranji L, Alkayyali T, Batun KD, Batman M, Toy B, Çiftaslan A. Growing burden of nonalcoholic fatty liver disease in Turkey: A single-center experience. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 30:892-898. [PMID: 31258138 DOI: 10.5152/tjg.2019.19072] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIMS Nonalcoholic fatty liver disease (NAFLD), which consists of nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH), is a growing epidemic in Turkey, considering the recent alarming prevalence of 48.3%. Patients with NASH and/or liver fibrosis are more likely to progress to advanced liver disease. In this single-center study, we sought to describe the clinical and histological characteristics of a sample of Turkish patients with biopsy-proven NAFLD, who were enrolled over a 4-year period. MATERIALS AND METHODS This is a retrospective analysis of prospectively collected data from a total of 468 patients (224 males, 244 females; median age, 47 [18-71]. The study cohort consisted of patients with biopsy-proven NAFLD who were followed up at our outpatient clinic from 2009 to 2010 and from 2017 to 2018. Histological classification of the biopsies was performed according to the Steatosis, Activity and Fibrosis (SAF) scoring allowing the use of Fatty Liver Inhibition of Progression (FLIP) algorithm and the NAFLD Activity Score (NAS) scoring system. RESULTS Based on the SAF scoring, most patients (90.4%) had biopsy-proven NASH, whereas the NAFL was much rarer (9.6%). The prevalence of significant fibrosis (≥F2), advanced fibrosis (≥F3), and cirrhosis (F=4) was 35.0%, 17.5%, and 3.8%, respectively. The percentage of lean, overweight, and obese patients with NAFLD was 6.4%, 32.6%, and 61%, respectively. Metabolic syndrome was prevalent in 63% of the patients and Type 2 diabetes mellitus in 33.5%. CONCLUSION The growing burden of NAFLD as a public health problem in Turkey is underscored by its marked histological severity in terms of NASH and fibrosis. Well-conducted clinical trials will be essential for slowing down the NASH progression.
Collapse
Affiliation(s)
- Yusuf Yılmaz
- Department of Gastroenterology, Marmara University School of Medicine, İstanbul, Turkey
| | - Haluk Tarık Kanı
- Department of Gastroenterology, Marmara University School of Medicine, İstanbul, Turkey
| | - Coşkun Özer Demirtaş
- Department of Gastroenterology, Marmara University School of Medicine, İstanbul, Turkey
| | - Eda Kaya
- İstanbul University-Cerrahpaşa, Cerrahpaşa School of Medicine, İstanbul, Turkey
| | | | | | | | | | - Mahmut Batman
- Marmara University School of Medicine, İstanbul, Turkey
| | - Berk Toy
- Marmara University School of Medicine, İstanbul, Turkey
| | | |
Collapse
|
237
|
Novel Combinatorial Regimen of Garcinol and Curcuminoids for Non-alcoholic Steatohepatitis (NASH) in Mice. Sci Rep 2020; 10:7440. [PMID: 32366854 PMCID: PMC7198554 DOI: 10.1038/s41598-020-64293-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 04/12/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive form of Non-alcoholic fatty liver disease (NAFLD), a chronic liver disease with a significant unmet clinical need. In this study, we examined the protective effects of Garcinia indica extract standardized to contain 20% w/w of Garcinol (GIE) and 95% Curcuminoids w/w from Curcuma longa (Curcuminoids) in a Stelic animal model (STAM) of NASH. The STAM mice developed steatosis, hepatocyte ballooning, and inflammation, which were significantly reduced by the combination of GIE and Curcuminoids, resulting in a lower NAFLD activity score. The treatment reduced fibrosis as observed by Sirius red staining, liver hydroxyproline content and mRNA levels of TGF- β and collagen in the liver. Immunostaining with alpha-smooth muscle actin (α SMA) revealed a significant reduction in hepatic stellate cells. Intriguingly, the combination regimen markedly decreased the mRNA levels of MCP1 and CRP and both mRNA and protein levels of TNF-α. NF-kB, reduced the hepatic and circulating FGF21 levels and altered the nonenzymatic (glutathione) and enzymatic antioxidant markers (Glutathione peroxidase, and superoxide dismutase). Our results suggest that the combination of GIE and Curcuminoids can reduce the severity of NASH by reducing steatosis, fibrosis, oxidative stress, and inflammation. The results suggest that the combinatorial regimen could be an effective supplement to prevent the progression of liver steatosis to inflammation and fibrosis in NASH.
Collapse
|
238
|
Alkayyali T, Qutranji L, Kaya E, Bakir A, Yilmaz Y. Clinical utility of noninvasive scores in assessing advanced hepatic fibrosis in patients with type 2 diabetes mellitus: a study in biopsy-proven non-alcoholic fatty liver disease. Acta Diabetol 2020; 57:613-618. [PMID: 31897769 DOI: 10.1007/s00592-019-01467-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM Simple noninvasive fibrosis scores based on routine blood tests have been increasingly investigated as screening tools in different clinical settings. Here, we sought to examine whether the Fibrosis-4 Index (FIB-4) and the non-alcoholic fatty liver disease (NAFLD) fibrosis score (NFS) could perform differently in diabetic versus non-diabetic patients with biopsy-proven NAFLD. METHODS We examined 349 patients with biopsy-proven NAFLD (166 with type 2 diabetes and 183 without). Patients with FIB-4 scores < 1.3 and > 2.67 or NFS scores < - 1.455 and > 0.676 were considered at low and high risk of advanced fibrosis, respectively. RESULTS A FIB-4 cutoff value of 1.3-which denotes a low risk of advanced fibrosis-had a specificity of 67% in patients with diabetes and 69% in those without. Conversely, a FIB-4 cutoff value of 2.67-which denotes a high risk of advanced fibrosis-had a sensitivity of 22% in patients with diabetes and 0% in those without. NFS performed similar to FIB-4. CONCLUSION Both FIB-4 and NFS scores have an acceptable clinical utility in the exclusion of advanced fibrosis in patients with NAFLD, regardless of the presence of type 2 diabetes. However, their usefulness in identifying advanced fibrosis is limited-especially in the absence of diabetes.
Collapse
Affiliation(s)
| | - Lubna Qutranji
- School of Medicine, Marmara University, Istanbul, Turkey
| | - Eda Kaya
- Institute of Gastroenterology, Marmara University, Istanbul, Turkey
| | - Alev Bakir
- Department of Biostatistics and Medical Informatics, School of Medicine, Halic University, Istanbul, Turkey
| | - Yusuf Yilmaz
- Institute of Gastroenterology, Marmara University, Istanbul, Turkey.
- Department of Gastroenterology, School of Medicine, Marmara University, Istanbul, Turkey.
| |
Collapse
|
239
|
Goldner D, Lavine JE. Nonalcoholic Fatty Liver Disease in Children: Unique Considerations and Challenges. Gastroenterology 2020; 158:1967-1983.e1. [PMID: 32201176 DOI: 10.1053/j.gastro.2020.01.048] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/30/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasing in prevalence in concert with the global epidemic of obesity and is being diagnosed at increasingly younger ages. The unique histologic features and early presentation of disease in pediatrics suggest that children and adults may differ with regard to etiopathogenesis, with children displaying a greater vulnerability to genetic and environmental factors. Of significant relevance to pediatrics, in utero and perinatal stressors may alter the lifelong health trajectory of a child, increasing the risk of NAFLD and other cardiometabolic diseases. The development and progression of disease in childhood is likely to carry increased risk of long-term morbidity. Novel biomarkers and therapeutic agents are needed to avoid the otherwise inevitable health and societal consequences of this rapidly expanding pediatric population.
Collapse
Affiliation(s)
- Dana Goldner
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York
| | - Joel E Lavine
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Columbia University Medical Center, New York, New York.
| |
Collapse
|
240
|
Johansen P, Howard D, Bishop R, Moreno SI, Buchholtz K. Systematic Literature Review and Critical Appraisal of Health Economic Models Used in Cost-Effectiveness Analyses in Non-Alcoholic Steatohepatitis: Potential for Improvements. PHARMACOECONOMICS 2020; 38:485-497. [PMID: 31919793 DOI: 10.1007/s40273-019-00881-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a severe, typically progressive form of non-alcoholic fatty liver disease (NAFLD). The global prevalence of NASH is increasing, driven partly by the global increase in obesity and type 2 diabetes mellitus (T2DM), such that NASH is now a leading cause of cirrhosis. There is currently an unmet clinical need for efficacious and cost-effective treatments for NASH; no pharmacologic agents have an approved indication for NASH. OBJECTIVE Our objective was to summarise and critically appraise published health economic models of NASH, to evaluate their quality and suitability for use in the assessment of novel treatments for NASH, and to identify knowledge gaps, challenges and opportunities for future modelling. METHODS A systematic literature review was performed using the MEDLINE, Embase, Cochrane Library and EconLit databases to identify published health economic analyses in patients with NAFLD or NASH. Supplementary hand searches of grey literature were also performed. Articles published up to November 2019 were included in the review. Quality assessment of identified studies was also performed. RESULTS A total of 19 articles comprising 16 unique models including either NAFLD as a whole or NASH alone were included in the review. Structurally, most models had a state-transition component; in terms of health states, two different approaches to early disease states were used, modelling either progression through fibrosis stages or NAFLD/NASH-specific health states. Conditions that frequently co-exist with NASH, such as obesity, T2DM and cardiovascular disease were not captured in models identified here. Late-stage complications such as cirrhosis, decompensated cirrhosis and hepatocellular carcinoma were consistently included, but input data (e.g. costs, utilities and transition probabilities) for late-stage complications were frequently sourced from other liver disease areas. The quality of included studies was heterogenous, and only a small proportion of studies reported internal and external validation processes. CONCLUSION The health economic models identified in this review are associated with limitations primarily driven by a lack of NASH-specific data. Identified models also largely overlooked the intricate association between NASH and other conditions, including obesity and T2DM, and did not capture the increased risk of cardiovascular events associated with NASH. High-quality, transparent, validated health economic models of NASH will be required to evaluate the cost effectiveness of treatments currently in development, particularly compounds that may target other non-hepatic outcomes.
Collapse
|
241
|
Reddy YK, Marella HK, Jiang Y, Ganguli S, Snell P, Podila PS, Maliakkal B, Satapathy SK. Natural History of Non-Alcoholic Fatty Liver Disease: A Study With Paired Liver Biopsies. J Clin Exp Hepatol 2020; 10:245-254. [PMID: 32405181 PMCID: PMC7212293 DOI: 10.1016/j.jceh.2019.07.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Although there is unequivocal evidence for progression of nonalcoholic steatohepatitis (NASH) to cirrhosis, there is uncertainty with regard to the progression to nonalcoholic fatty liver (NAFL) and NASH. AIMS We investigated the rate of progression to NASH and advanced fibrosis in patients with nonalcoholic fatty liver disease (NAFLD) and assessed the factors associated with such progression. METHODS Histological assessment was performed in 36 patients with NAFLD with paired liver biopsies (≥1 year apart; median, 3.8 years; range, 1-10.33 years). NASH Clinical Research Network (NASH CRN) criteria were used to assess NAFLD Activity Score (NAS). RESULTS At baseline, 26 (72%) patients had NAFL and 10 (28%) patients had NASH. At follow-up, 27% NAFL progressed to NASH (NAS score ≥5), and 50% of patients with NASH no longer met the criteria of NASH. Fibrosis progressed in 15 (42%), regressed in 9 (25%), and remained stable in 12 (33%) patients overall. Thirty-five percent of patients with NAFL had fibrosis progression. The incidence of type 2 diabetes mellitus (T2DM) was higher in patients with NASH versus NAFL (40% vs. 27%). Both at the time of baseline and follow-up, liver biopsies, composite models of noninvasive scores such as Fibrosis-4 (FIB-4) score and NAFLD fibrosis score, and ratio of aspartate aminotransferase (AST) to alanine aminotransferase (ALT) were all significantly higher in progressors than in nonprogressors. CONCLUSIONS NAFLD is a dynamic liver disease with varying degrees of progression and regression. T2DM was strongly associated with fibrosis progression. Noninvasive fibrosis scores such as AST/ALT ratio, FIB-4 score, and NAFLD fibrosis score can identify those at risk of fibrosis progression.
Collapse
Affiliation(s)
- Yala K. Reddy
- Division of Transplant Surgery, Department of Surgery, Methodist University Hospital Transplant Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38104, USA
| | - Hemnishil K. Marella
- Division of Transplant Surgery, Department of Surgery, Methodist University Hospital Transplant Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38104, USA
| | - Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental Health, University of Memphis, Memphis, TN, 38152, USA
| | - Surosree Ganguli
- Department of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Peter Snell
- Division of Transplant Surgery, Department of Surgery, Methodist University Hospital Transplant Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38104, USA
| | - Pradeep S.B. Podila
- Methodist University Hospital, Methodist Le Bonheur Healthcare, Memphis, TN, USA
| | - Benedict Maliakkal
- Division of Transplant Surgery, Department of Surgery, Methodist University Hospital Transplant Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38104, USA
| | - Sanjaya K. Satapathy
- Division of Hepatology and Sandra Atlas Bass Center for Liver Diseases, Donald and Barbara Zucker School of Medicine/Northwell Health, Manhasset, NY, 11030, USA
| |
Collapse
|
242
|
Cotter TG, Rinella M. Nonalcoholic Fatty Liver Disease 2020: The State of the Disease. Gastroenterology 2020; 158:1851-1864. [PMID: 32061595 DOI: 10.1053/j.gastro.2020.01.052] [Citation(s) in RCA: 812] [Impact Index Per Article: 162.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/09/2020] [Accepted: 01/11/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease, with a worldwide prevalence of 25%. In the United States, NAFLD and its subtype, nonalcoholic steatohepatitis, affect 30% and 5% of the population, respectively. Considering the ongoing obesity epidemic beginning in childhood, the rise in diabetes, and other factors, the prevalence of NAFLD along with the proportion of those with advanced liver disease is projected to continue to increase. This will have an important impact on public health reflected in health care costs, including impact on the need for liver transplantation, for which nonalcoholic steatohepatitis is already close to becoming the most common indication. NAFLD patients with evidence of nonalcoholic steatohepatitis and advanced fibrosis are at markedly increased risk of adverse outcomes, including overall mortality, and liver-specific morbidity and mortality, respectively. Identification of this cohort of NAFLD patients is paramount, given the associated poorer outcomes, in order to target resources to those who need it most. Various noninvasive tools have been developed in this regard. This review provides an update on the epidemiology, clinical and prognostic features, and diagnostic approach to patients with NAFLD.
Collapse
Affiliation(s)
- Thomas G Cotter
- Division of Gastroenterology and Hepatology, The University of Chicago Medicine, Chicago, Illinois
| | - Mary Rinella
- Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
243
|
Mitchell MC, Kerr T, Herlong HF. Current Management and Future Treatment of Alcoholic Hepatitis. Gastroenterol Hepatol (N Y) 2020; 16:178-189. [PMID: 34035720 PMCID: PMC8132686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Excessive alcohol consumption is responsible for approximately 50% of all deaths due to cirrhosis. Although the duration and amount of alcohol consumption are the primary factors responsible for the liver injury caused by consuming alcohol, the pathogenesis of the 3 stages of alcohol-associated liver disease (ALD)-fatty liver, alcoholic hepatitis (AH), and cirrhosis- is likely multifactorial. Preexisting obesity, dysbiosis of the gut microbiome, activation of proinflammatory cytokines, and genetic factors can all contribute to the risk of developing ALD. The cornerstone of therapy for all stages of ALD is abstinence from drinking alcoholic beverages. Severe AH, defined by a Maddrey discriminant function greater than 32, warrants additional therapy. The results of multiple studies evaluating the use of glucocorticoids in the treatment of severe AH led to guidelines from international societies that recommend glucocorticoid therapy in patients with severe AH without active infection. Liver transplantation provides an effective treatment option for patients who fail glucocorticoid therapy. Recent advances in understanding the pathogenesis of AH have led to the investigation of potential therapies directed at preventing the development of steatosis, inhibiting the innate immune response, modifying the gut microbiome, and stimulating liver regeneration.
Collapse
Affiliation(s)
- Mack C Mitchell
- Dr Mitchell is the Nancy S. and Jeremy L. Halbreich Professor of Gastroenterology and vice president of medical affairs at the University of Texas Southwestern Medical Center in Dallas, Texas. Dr Kerr is an associate professor and Dr Herlong is a professor in the Division of Digestive and Liver Diseases at the University of Texas Southwestern Medical Center
| | - Thomas Kerr
- Dr Mitchell is the Nancy S. and Jeremy L. Halbreich Professor of Gastroenterology and vice president of medical affairs at the University of Texas Southwestern Medical Center in Dallas, Texas. Dr Kerr is an associate professor and Dr Herlong is a professor in the Division of Digestive and Liver Diseases at the University of Texas Southwestern Medical Center
| | - H Franklin Herlong
- Dr Mitchell is the Nancy S. and Jeremy L. Halbreich Professor of Gastroenterology and vice president of medical affairs at the University of Texas Southwestern Medical Center in Dallas, Texas. Dr Kerr is an associate professor and Dr Herlong is a professor in the Division of Digestive and Liver Diseases at the University of Texas Southwestern Medical Center
| |
Collapse
|
244
|
Kamata Y, Kessoku T, Shimizu T, Kobayashi T, Kurihashi T, Sato S, Kuraji S, Aoyama N, Iwasaki T, Takashiba S, Hamada N, Kodama T, Tamura T, Ino S, Higurashi T, Taguri M, Yamanaka T, Yoneda M, Usuda H, Wada K, Nakajima A, Minabe M. Efficacy and safety of PERIOdontal treatment versus usual care for Nonalcoholic liver disease: protocol of the PERION multicenter, two-arm, open-label, randomized trial. Trials 2020; 21:291. [PMID: 32293522 PMCID: PMC7092586 DOI: 10.1186/s13063-020-4201-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background We report the first protocol for a multicenter, randomized comparison study to compare the efficacies of periodontal scaling and root-planing treatment against that of tooth-brushing treatment for nonalcoholic fatty liver disease (NAFLD) (PERION: PERIOdontal treatment for NAFLD). Nonalcoholic steatohepatitis (NASH) is an advanced form of NAFLD, which can progress to cirrhosis and hepatocellular carcinoma. Increased endotoxemia is associated with the progression of NAFLD. Periodontal bacteria possess endotoxins; Porphyromonas gingivalis is well-known as a major pathogenic bacterium in periodontitis, and serum antibody levels for P. gingivalis are high in patients with periodontitis. Several reports have indicated that P. gingivalis is related to NAFLD. This study aims to investigate the effect of periodontal treatment for liver damage, P. gingivalis infection, and endotoxemia on patients with NAFLD. Methods We will include adult patients (20–85 years old) with NAFLD, alanine aminotransferase (ALT) ≥ 40 IU/L, and equivalent steatosis grade ≥ 1 (target sample size, n = 40 patients; planned number of patients with outcome data, n = 32). Participants will be randomly assigned to one of two groups: a scaling and root-planing group or tooth-brushing as the usual group. The primary outcome will be the change in ALT levels from baseline to 12 weeks; the key secondary outcome will be the change in the serum immunoglobulin G (IgG) antibody titer for P. gingivalis at 12 weeks. Discussion This study should determine whether periodontal treatment decreases liver damage, P. gingivalis infection, and endotoxemia in patients with NAFLD. Trial registration University Hospital Medical Information Network (UMIN) Clinical Trials Registry, ID: UMIN000022079.
Collapse
Affiliation(s)
- Yohei Kamata
- Department of Highly Advanced Oral Stomatology, Yokohama Clinic, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Tomoko Shimizu
- Department of Highly Advanced Oral Stomatology, Yokohama Clinic, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takeo Kurihashi
- Department of Internal Medicine, Yokohama Clinic, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Satsuki Sato
- Department of Highly Advanced Oral Stomatology, Yokohama Clinic, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Syotaro Kuraji
- Department of Highly Advanced Oral Stomatology, Yokohama Clinic, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Norio Aoyama
- Division of Periodontology, Department of Oral Interdisciplinary Medicine, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa, 238-8580, Japan
| | - Tomoyuki Iwasaki
- Iwasaki Internal Medicine Clinic, 1-1-5 Furu-ruyokohama1F, Kamihoshikawa, Hodogaya-ku Yokohama, Kanagawa, 240-0042, Japan
| | - Shogo Takashiba
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Nobushiro Hamada
- Division of Microbiology, Department of Oral Science Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa, 238-8580, Japan
| | - Toshiro Kodama
- Department of Implantology and Periodontology, Graduate School of Dentistry, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Toshiyuki Tamura
- Department of Highly Advanced Oral Stomatology, Yokohama Clinic, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Satoshi Ino
- Division of Prosthetic Dentistry, Department of Highly Advanced Stomatology, Graduate School of Dentistry, Kanagawa Dental University, 3-31-6 Tsuruya-cho, Kanagawa, Yokohama, Kanagawa, 221-0835, Japan
| | - Takuma Higurashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Masataka Taguri
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Haruki Usuda
- Department of Pharmacology, Shimane University School of Medicine, 89-1 Enya-cho Izumo, Shimane, 693-0581, Japan
| | - Koichiro Wada
- Department of Pharmacology, Shimane University School of Medicine, 89-1 Enya-cho Izumo, Shimane, 693-0581, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Masato Minabe
- Division of Periodontology, Department of Oral Interdisciplinary Medicine, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa, 238-8580, Japan.
| |
Collapse
|
245
|
Ocker M. Challenges and opportunities in drug development for nonalcoholic steatohepatitis. Eur J Pharmacol 2020; 870:172913. [PMID: 31926994 DOI: 10.1016/j.ejphar.2020.172913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/04/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are considered major global medical burdens with high prevalence and steeply rising incidence. Despite the characterization of numerous pathophysiologic pathways leading to metabolic disorder, lipid accumulation, inflammation, fibrosis, and ultimately end-stage liver disease or liver cancer formation, so far no causal pharmacological therapy is available. Drug development for NAFLD and NASH is limited by long disease duration and slow progression and the need for sequential biopsies to monitor the disease stage. Additional non-invasive biomarkers could therefore improve design and feasibility of such. Here, the current concepts on preclinical models, biomarkers and clinical endpoints and trial designs are briefly reviewed.
Collapse
|
246
|
Hernandez GV, Smith VA, Melnyk M, Burd MA, Sprayberry KA, Edwards MS, Peterson DG, Bennet DC, Fanter RK, Columbus DA, Steibel JP, Glanz H, Immoos C, Rice MS, Santiago-Rodriguez TM, Blank J, VanderKelen JJ, Kitts CL, Piccolo BD, La Frano MR, Burrin DG, Maj M, Manjarin R. Dysregulated FXR-FGF19 signaling and choline metabolism are associated with gut dysbiosis and hyperplasia in a novel pig model of pediatric NASH. Am J Physiol Gastrointest Liver Physiol 2020; 318:G582-G609. [PMID: 32003601 PMCID: PMC7099491 DOI: 10.1152/ajpgi.00344.2019] [Citation(s) in RCA: 25] [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: 11/13/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 02/08/2023]
Abstract
To investigate the role of bile acids (BAs) in the pathogenesis of diet-induced nonalcoholic steatohepatitis (NASH), we fed a "Western-style diet" [high fructose, high fat (HFF)] enriched with fructose, cholesterol, and saturated fat for 10 wk to juvenile Iberian pigs. We also supplemented probiotics with in vitro BA deconjugating activity to evaluate their potential therapeutic effect in NASH. Liver lipid and function, cytokines, and hormones were analyzed using commercially available kits. Metabolites, BAs, and fatty acids were measured by liquid chromatography-mass spectrometry. Histology and gene and protein expression analyses were performed using standard protocols. HFF-fed pigs developed NASH, cholestasis, and impaired enterohepatic Farnesoid-X receptor (FXR)-fibroblast growth factor 19 (FGF19) signaling in the absence of obesity and insulin resistance. Choline depletion in HFF livers was associated with decreased lipoprotein and cholesterol in serum and an increase of choline-containing phospholipids in colon contents and trimethylamine-N-oxide in the liver. Additionally, gut dysbiosis and hyperplasia increased with the severity of NASH, and were correlated with increased colonic levels of choline metabolites and secondary BAs. Supplementation of probiotics in the HFF diet enhanced NASH, inhibited hepatic autophagy, increased excretion of taurine and choline, and decreased gut microbial diversity. In conclusion, dysregulation of BA homeostasis was associated with injury and choline depletion in the liver, as well as increased biliary secretion, gut metabolism and excretion of choline-based phospholipids. Choline depletion limited lipoprotein synthesis, resulting in hepatic steatosis, whereas secondary BAs and choline-containing phospholipids in colon may have promoted dysbiosis, hyperplasia, and trimethylamine synthesis, causing further damage to the liver.NEW & NOTEWORTHY Impaired Farnesoid-X receptor (FXR)-fibroblast growth factor 19 (FGF19) signaling and cholestasis has been described in nonalcoholic fatty liver disease (NAFLD) patients. However, therapeutic interventions with FXR agonists have produced contradictory results. In a swine model of pediatric nonalcoholic steatohepatitis (NASH), we show that the uncoupling of intestinal FXR-FGF19 signaling and a decrease in FGF19 levels are associated with a choline-deficient phenotype of NASH and increased choline excretion in the gut, with the subsequent dysbiosis, colonic hyperplasia, and accumulation of trimethylamine-N-oxide in the liver.
Collapse
Affiliation(s)
- Gabriella V Hernandez
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Victoria A Smith
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Megan Melnyk
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
| | - Matthew A Burd
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Kimberly A Sprayberry
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Mark S Edwards
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Daniel G Peterson
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Darin C Bennet
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| | - Rob K Fanter
- Center for Health Research, California Polytechnic State University, San Luis Obispo, California
| | | | - Juan P Steibel
- Department of Animal Science and Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan
| | - Hunter Glanz
- Department of Statistics, California Polytechnic State University, San Luis Obispo, California
| | - Chad Immoos
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California
| | - Margaret S Rice
- Department of Chemistry and Biochemistry, California Polytechnic State University, San Luis Obispo, California
| | | | - Jason Blank
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
| | - Jennifer J VanderKelen
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| | - Christopher L Kitts
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| | - Brian D Piccolo
- United States Department of Agriculture-Agricultural Research Services, Arkansas Children's Nutrition Center, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Michael R La Frano
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, California
- Center for Health Research, California Polytechnic State University, San Luis Obispo, California
| | - Douglas G Burrin
- United States Department of Agriculture-Agricultural Research Services, Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Magdalena Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, California
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, California
| | - Rodrigo Manjarin
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, California
| |
Collapse
|
247
|
Okamoto K, Koda M, Okamoto T, Onoyama T, Miyoshi K, Kishina M, Matono T, Kato J, Tokunaga S, Sugihara T, Hiramatsu A, Hyogo H, Tobita H, Sato S, Kawanaka M, Hara Y, Hino K, Chayama K, Murawaki Y, Isomoto H. Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease. PLoS One 2020; 15:e0219412. [PMID: 32106257 PMCID: PMC7046274 DOI: 10.1371/journal.pone.0219412] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/10/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) has a wide spectrum, eventually leading to cirrhosis and hepatic carcinogenesis. We previously reported that a series of microRNAs (miRNAs) mapped in the 14q32.2 maternally imprinted gene region (Dlk1-Dio3 mat) are related to NAFLD development and progression in a mouse model. We examined the suitability of miR-379, a circulating Dlk1-Dio3 mat miRNA, as a human NAFLD biomarker. Methods Eighty NAFLD patients were recruited for this study. miR-379 was selected from the putative Dlk1-Dio3 mat miRNA cluster because it exhibited the greatest expression difference between NAFLD and non-alcoholic steatohepatitis in our preliminary study. Real-time PCR was used to examine the expression levels of miR-379 and miR-16 as an internal control. One patient was excluded due to low RT-PCR signal. Results Compared to normal controls, serum miR-379 expression was significantly up-regulated in NAFLD patients. Receiver operating characteristic curve analysis suggested that miR-379 is a suitable marker for discriminating NAFLD patients from controls, with an area under the curve value of 0.72. Serum miR-379 exhibited positive correlations with alkaline phosphatase, total cholesterol, low-density-lipoprotein cholesterol and non-high-density-lipoprotein cholesterol levels in patients with early stage NAFLD (Brunt fibrosis stage 0 to 1). The correlation between serum miR-379 and cholesterol levels was lost in early stage NAFLD patients treated with statins. Software-based predictions indicated that various energy metabolism–related genes, including insulin-like growth factor-1 (IGF-1) and IGF-1 receptor, are potential targets of miR-379. Conclusions Serum miR-379 exhibits high potential as a biomarker for NAFLD. miR-379 appears to increase cholesterol lipotoxicity, leading to the development and progression of NAFLD, via interference with the expression of target genes, including those related to the IGF-1 signaling pathway. Our results could facilitate future research into the pathogenesis, diagnosis, and treatment of NAFLD.
Collapse
Affiliation(s)
- Kinya Okamoto
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
- * E-mail:
| | - Masahiko Koda
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Toshiaki Okamoto
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Takumi Onoyama
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Kenichi Miyoshi
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Manabu Kishina
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Tomomitsu Matono
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Jun Kato
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Shiho Tokunaga
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Takaaki Sugihara
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Akira Hiramatsu
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Hideyuki Hyogo
- Department of Gastroenterology and Hepatology, JA Hiroshima General Hospital, Hatsukaichi, Hiroshima, Japan
| | - Hiroshi Tobita
- Department of Gastroenterology and Hepatology, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Shuichi Sato
- Department of Gastroenterology and Hepatology, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, General Medical Center, Kawasaki Medical School, Okayama, Okayama, Japan
| | - Yuichi Hara
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Yoshikazu Murawaki
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Hajime Isomoto
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
248
|
Zhang R, Chu K, Zhao N, Wu J, Ma L, Zhu C, Chen X, Wei G, Liao M. Corilagin Alleviates Nonalcoholic Fatty Liver Disease in High-Fat Diet-Induced C57BL/6 Mice by Ameliorating Oxidative Stress and Restoring Autophagic Flux. Front Pharmacol 2020; 10:1693. [PMID: 32116684 PMCID: PMC7011087 DOI: 10.3389/fphar.2019.01693] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022] Open
Abstract
Corilagin (Cori) possesses multiple biological activities. To determine whether Cori can exert protective effects against nonalcoholic fatty liver disease (NAFLD) and its potential mechanisms. C57BL/6 mice were fed with high-fat diet (HFD) alone or in combination with Cori (20 mg/kg, i.p.) and AML12 cells were exposed to 200 μM PA/OA with or without Cori (10 μM or 20 μM). Phenotypes and key indicators relevant to NAFLD were examined both in vivo and in vitro. In this study, Cori significantly ameliorated hepatic steatosis, confirmed by improved serum lipid profiles, and hepatic TC, TG contents, and the gene expression related to lipid metabolism in livers of HFD mice. Moreover, Cori attenuated HFD-mediated autophagy (including mitophagy) blockage by restoring autophagic flux, evidenced by increased number of autophagic double vesicles containing mitochondria, elevated LC3II protein levels, decreased p62 protein levels, as well as enhanced colocalization of autophagy-related protein (LC3, Parkin) and mitochondria. In accordance with this, Cori also reduced the accumulation of ROS and MDA levels, and enhanced the activities of antioxidative enzymes including SOD, GSH-Px, and CAT. In addition, Cori treatment improved mitochondrial dysfunction, evidenced by increased mitochondrial membrane potential (ΔΨm). In parallel with this, Cori decreased mitochondrial DNA oxidative damage, while increased mitochondrial biogenesis related transcription factors expression, mitochondrial DNA content and oxygen consumption rate (OCR). In conclusion, these results demonstrate that Cori is a potential candidate for the treatment of NAFLD via diminishing oxidative stress, restoring autophagic flux, as well as improving mitochondrial functions.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kexin Chu
- Department of Radiation Oncology, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Nengjiang Zhao
- Department of Traditional Chinese Medicine Studio, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jingjing Wu
- Department of Breast, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lina Ma
- Department of Breast, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenfang Zhu
- Department of General Surgery, The Ninth People's Hospital, Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Xia Chen
- Department of Endocrinology and Metabolism, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Gang Wei
- Department of Endocrinology and Metabolism, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Diabetes, Shanghai Institute for Diabetes, Shanghai Clinical Medical Centre of Diabetes, Shanghai Key Clinical Centre of Metabolic Diseases, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mingjuan Liao
- Department of Traditional Chinese Medicine, The Ninth People's Hospital, Medical School of Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
249
|
Bril F, McPhaul MJ, Caulfield MP, Clark VC, Soldevilla-Pico C, Firpi-Morell RJ, Lai J, Shiffman D, Rowland CM, Cusi K. Performance of Plasma Biomarkers and Diagnostic Panels for Nonalcoholic Steatohepatitis and Advanced Fibrosis in Patients With Type 2 Diabetes. Diabetes Care 2020; 43:290-297. [PMID: 31604692 DOI: 10.2337/dc19-1071] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/10/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The 2019 Standards of Medical Care in Diabetes suggested that patients with nonalcoholic fatty liver disease (NAFLD) should be evaluated for liver fibrosis. However, the performance of noninvasive clinical models/scores and plasma biomarkers for the diagnosis of nonalcoholic steatohepatitis (NASH) and advanced fibrosis has not been carefully assessed in patients with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS In this cross-sectional study, patients (n = 213) had a liver MRS, and those with a diagnosis of NAFLD underwent a percutaneous liver biopsy. Several noninvasive clinical models/scores and plasma biomarkers were measured to identify NASH and advanced fibrosis (NASH: ALT, cytokeratin-18, NashTest 2, HAIR, BARD, and OWLiver; advanced fibrosis: AST, fragments of propeptide of type III procollagen [PRO-C3], FIB-4, APRI, NAFLD fibrosis score, and FibroTest). RESULTS None of the noninvasive tools assessed for the diagnosis of NASH in patients with T2DM had an optimum performance (all areas under the curve [AUCs] <0.80). Of note, none of the panels or biomarkers was able to outperform plasma ALT (AUC 0.78 [95% CI 0.71-0.84]). Performance was better to diagnose advanced fibrosis, in which plasma PRO-C3, AST, and APRI showed better results than the other approaches (AUC 0.90 [0.85-0.95], 0.85 [0.80-0.91], and 0.86 [0.80-0.91], respectively). Again, none of the approaches did significantly better than plasma AST. Sequential use of plasma AST and other noninvasive tests may help in limiting the number of liver biopsies required to identify patients with advanced fibrosis. CONCLUSIONS Performance of noninvasive clinical models/scores and plasma biomarkers for the diagnosis of NASH or advanced fibrosis was suboptimal in patients with T2DM. Combination of multiple tests may provide an alternative to minimize the need for liver biopsies to detect fibrosis in these patients.
Collapse
Affiliation(s)
- Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL
- Internal Medicine, Department of Medicine, University of Florida, Gainesville, FL
| | | | | | - Virginia C Clark
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, FL
| | | | - Roberto J Firpi-Morell
- Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, FL
| | - Jinping Lai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Dov Shiffman
- Quest Diagnostics Nichols Institute, San Juan Capistrano, CA
| | | | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL
- Division of Endocrinology, Diabetes and Metabolism, Malcom Randall VA Medical Center, Gainesville, FL
| |
Collapse
|
250
|
Anstee QM, Neuschwander-Tetri BA, Wong VWS, Abdelmalek MF, Younossi ZM, Yuan J, Pecoraro ML, Seyedkazemi S, Fischer L, Bedossa P, Goodman Z, Alkhouri N, Tacke F, Sanyal A. Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: AURORA Phase 3 study design. Contemp Clin Trials 2020; 89:105922. [PMID: 31881392 DOI: 10.1016/j.cct.2019.105922] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nonalcoholic steatohepatitis (NASH) is a sub-classification of nonalcoholic fatty liver disease (NAFLD) characterized by increased risk of progressive liver fibrosis. Cenicriviroc (CVC) is a novel, orally administered, potent chemokine 2 and 5 receptor antagonist currently in development for the treatment of liver fibrosis in adults with NASH. METHODS AND ANALYSIS Efficacy and safety of CVC will be comprehensively evaluated in a global, Phase 3, multicenter, randomized, double-blind, placebo-controlled study (AURORA, NCT03028740) of subjects with NASH and Stage F2 or F3 fibrosis. Approximately 2000 adults (Part 1, 1200 subjects; Part 2, 800 additional subjects) aged 18-75 years with histological evidence of NASH with Stage F2 or F3 fibrosis (NASH Clinical Research Network classification system) will be randomized 2:1 to CVC 150 mg or placebo orally once daily. Primary efficacy endpoints will include the proportion of subjects with ≥1-stage improvement in liver fibrosis and no worsening of steatohepatitis at Month 12 relative to screening (Part 1), and time to first occurrence of any adjudicated event: death; histopathologic progression to cirrhosis; liver transplant; Model of End-Stage Liver Disease score ≥ 15; ascites; hospitalization due to liver decompensation (Part 2). Patient-reported outcomes will assess changes in health outcomes from baseline (Chronic Liver Disease Questionnaire - NAFLD; Work Productivity and Activity Impairment in NASH; 36-Item Short Form Health Survey version 2). Adverse events will be assessed throughout the study. As there are currently no approved treatments indicated for NASH, the AURORA CVC Phase 3 study addresses an unmet medical need.
Collapse
Affiliation(s)
- Quentin M Anstee
- Institute of Translational & Clinical Research, 4(th) floor, William Leech Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| | - Brent A Neuschwander-Tetri
- Division of Gastroenterology and Hepatology, Saint Louis University, 3635 Vista Avenue, St. Louis, MO 63110, USA
| | - Vincent Wai-Sun Wong
- Chinese University of Hong Kong, Department of Medicine and Therapeutics, 9/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | | | - Zobair M Younossi
- Center for Liver Diseases and Department of Medicine, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Jiacheng Yuan
- Allergan plc, 2525 Dupont Drive, Irvine, CA 92612, USA
| | | | - Star Seyedkazemi
- Allergan plc, 701 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Laurent Fischer
- Allergan plc, 701 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Pierre Bedossa
- Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110 Clichy, France
| | - Zachary Goodman
- Center for Liver Diseases and Department of Medicine, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Naim Alkhouri
- Texas Liver Institute, University of Texas Health Science Center, 607 Camden Street, Suite 108, San Antonio, TX 78215, USA
| | - Frank Tacke
- Charité University Medical Center Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Arun Sanyal
- Virginia Commonwealth University, 1201 East Marshall Street, Richmond, VA 23298, USA
| |
Collapse
|