51
|
Nakao Y, Fukushima M, Mauer AS, Liao CY, Ferris A, Dasgupta D, Heppelmann CJ, Vanderboom PM, Saraswat M, Pandey A, Nair KS, Allen AM, Nakao K, Malhi H. A Comparative Proteomic Analysis of Extracellular Vesicles Associated With Lipotoxicity. Front Cell Dev Biol 2021; 9:735001. [PMID: 34805145 PMCID: PMC8600144 DOI: 10.3389/fcell.2021.735001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging mediators of intercellular communication in nonalcoholic steatohepatitis (NASH). Palmitate, a lipotoxic saturated fatty acid, activates hepatocellular endoplasmic reticulum stress, which has been demonstrated to be important in NASH pathogenesis, including in the release of EVs. We have previously demonstrated that the release of palmitate-stimulated EVs is dependent on the de novo synthesis of ceramide, which is trafficked by the ceramide transport protein, STARD11. The trafficking of ceramide is a critical step in the release of lipotoxic EVs, as cells deficient in STARD11 do not release palmitate-stimulated EVs. Here, we examined the hypothesis that protein cargoes are trafficked to lipotoxic EVs in a ceramide-dependent manner. We performed quantitative proteomic analysis of palmitate-stimulated EVs in control and STARD11 knockout hepatocyte cell lines. Proteomics was performed on EVs isolated by size exclusion chromatography, ultracentrifugation, and density gradient separation, and EV proteins were measured by mass spectrometry. We also performed human EV proteomics from a control and a NASH plasma sample, for comparative analyses with hepatocyte-derived lipotoxic EVs. Size exclusion chromatography yielded most unique EV proteins. Ceramide-dependent lipotoxic EVs contain damage-associated molecular patterns and adhesion molecules. Haptoglobin, vascular non-inflammatory molecule-1, and insulin-like growth factor-binding protein complex acid labile subunit were commonly detected in NASH and hepatocyte-derived ceramide-dependent EVs. Lipotoxic EV proteomics provides novel candidate proteins to investigate in NASH pathogenesis and as diagnostic biomarkers for hepatocyte-derived EVs in NASH patients.
Collapse
Affiliation(s)
- Yasuhiko Nakao
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Masanori Fukushima
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Amy S Mauer
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| | - Chieh-Yu Liao
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| | - Anya Ferris
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,California Polytechnic State University, San Luis Obispo, CA, United States
| | - Debanjali Dasgupta
- Division of Gastroenterology and Hepatology, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Manipal, India
| | | | - Patrick M Vanderboom
- Mayo Clinic Medical Genome Facility-Proteomics Core, Manipal, India.,Mayo Endocrine Research Unit, Manipal, India
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States.,Institute of Bioinformatics, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States.,Institute of Bioinformatics, Bangalore, India.,Manipal Academy of Higher Education (MAHE), Manipal, India.,Center for Individualized Medicine, Rochester, MN, United States
| | | | - Alina M Allen
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Rochester, MN, United States
| |
Collapse
|
52
|
Kostallari E, Valainathan S, Biquard L, Shah VH, Rautou PE. Role of extracellular vesicles in liver diseases and their therapeutic potential. Adv Drug Deliv Rev 2021; 175:113816. [PMID: 34087329 PMCID: PMC10798367 DOI: 10.1016/j.addr.2021.05.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/17/2021] [Accepted: 05/29/2021] [Indexed: 02/07/2023]
Abstract
More than eight hundred million people worldwide have chronic liver disease, with two million deaths per year. Recurring liver injury results in fibrogenesis, progressing towards cirrhosis, for which there doesn't exists any cure except liver transplantation. Better understanding of the mechanisms leading to cirrhosis and its complications is needed to develop effective therapies. Extracellular vesicles (EVs) are released by cells and are important for cell-to-cell communication. EVs have been reported to be involved in homeostasis maintenance, as well as in liver diseases. In this review, we present current knowledge on the role of EVs in non-alcoholic fatty liver disease and non-alcoholic steatohepatitis, alcohol-associated liver disease, chronic viral hepatitis, primary liver cancers, acute liver injury and liver regeneration. Moreover, therapeutic strategies involving EVs as targets or as tools to treat liver diseases are summarized.
Collapse
Affiliation(s)
- Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States.
| | - Shantha Valainathan
- Université de Paris, AP-HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Centre de recherche sur l'inflammation, Inserm, UMR 1149, Paris, France
| | - Louise Biquard
- Université de Paris, Centre de recherche sur l'inflammation, Inserm, UMR 1149, Paris, France.
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States.
| | - Pierre-Emmanuel Rautou
- Université de Paris, AP-HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Centre de recherche sur l'inflammation, Inserm, UMR 1149, Paris, France.
| |
Collapse
|
53
|
Donoso‐Quezada J, Ayala‐Mar S, González‐Valdez J. The role of lipids in exosome biology and intercellular communication: Function, analytics and applications. Traffic 2021; 22:204-220. [PMID: 34053166 PMCID: PMC8361711 DOI: 10.1111/tra.12803] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/18/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Exosomes are extracellular vesicles that in recent years have received special attention for their regulatory functions in numerous biological processes. Recent evidence suggests a correlation between the composition of exosomes in body fluids and the progression of some disorders, such as cancer, diabetes and neurodegenerative diseases. In consequence, numerous studies have been performed to evaluate the composition of these vesicles, aiming to develop new biomarkers for diagnosis and to find novel therapeutic targets. On their part, lipids represent one of the most important components of exosomes, with important structural and regulatory functions during exosome biogenesis, release, targeting and cellular uptake. Therefore, exosome lipidomics has emerged as an innovative discipline for the discovery of novel lipid species with biomedical applications. This review summarizes the current knowledge about exosome lipids and their roles in exosome biology and intercellular communication. Furthermore, it presents the state-of-the-art analytical procedures used in exosome lipidomics while emphasizing how this emerging discipline is providing new insights for future applications of exosome lipids in biomedicine.
Collapse
Affiliation(s)
| | - Sergio Ayala‐Mar
- Tecnologico de MonterreySchool of Engineering and ScienceMonterreyNuevo LeónMexico
| | - José González‐Valdez
- Tecnologico de MonterreySchool of Engineering and ScienceMonterreyNuevo LeónMexico
| |
Collapse
|
54
|
Lee YT, Tran BV, Wang JJ, Liang IY, You S, Zhu Y, Agopian VG, Tseng HR, Yang JD. The Role of Extracellular Vesicles in Disease Progression and Detection of Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:3076. [PMID: 34203086 PMCID: PMC8233859 DOI: 10.3390/cancers13123076] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and one of the leading causes of cancer-related death worldwide. Despite the improvements in surveillance and treatment, the prognosis of HCC remains poor. Extracellular vesicles (EVs) are a heterogeneous group of phospholipid bilayer-enclosed particles circulating in the bloodstream and mediating intercellular communication. Emerging studies have shown that EVs play a crucial role in regulating the proliferation, immune escape, and metastasis of HCC. In addition, because EVs are present in the circulation at relatively early stages of disease, they are getting attention as an attractive biomarker for HCC detection. Over the past decade, dedicated efforts have been made to isolate EVs more efficiently and make them useful tools in different clinical settings. In this review article, we provide an overview of the EVs isolation methods and highlight the role of EVs as mediators in the pathogenesis and progression of HCC. Lastly, we summarize the potential applications of EVs in early-stage HCC detection.
Collapse
Affiliation(s)
- Yi-Te Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA 90095, USA; (Y.-T.L.); (I.Y.L.); (Y.Z.); (H.-R.T.)
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA;
| | - Benjamin V. Tran
- Department of Surgery, University of California, Los Angeles, CA 90095, USA; (B.V.T.); (V.G.A.)
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90048, USA
| | - Jasmine J. Wang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA;
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Icy Y. Liang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA 90095, USA; (Y.-T.L.); (I.Y.L.); (Y.Z.); (H.-R.T.)
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA 90095, USA; (Y.-T.L.); (I.Y.L.); (Y.Z.); (H.-R.T.)
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA;
| | - Vatche G. Agopian
- Department of Surgery, University of California, Los Angeles, CA 90095, USA; (B.V.T.); (V.G.A.)
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90048, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, CA 90095, USA; (Y.-T.L.); (I.Y.L.); (Y.Z.); (H.-R.T.)
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA;
| | - Ju Dong Yang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Comprehensive Transplant Center Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
55
|
He L, Sehrawat TS, Verma VK, Navarro-Corcuera A, Sidhu G, Mauer A, Luo X, Katsumi T, Chen J, Shah S, Arab JP, Cao S, Kashkar H, Gores GJ, Malhi H, Shah VH. XIAP Knockdown in Alcohol-Associated Liver Disease Models Exhibits Divergent in vitro and in vivo Phenotypes Owing to a Potential Zonal Inhibitory Role of SMAC. Front Physiol 2021; 12:664222. [PMID: 34025452 PMCID: PMC8138467 DOI: 10.3389/fphys.2021.664222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/31/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol-associated liver disease (ALD) has been recognized as the most common cause of advanced liver disease worldwide, though mechanisms of pathogenesis remain incompletely understood. The X-linked inhibitor of apoptosis (XIAP) protein was originally described as an anti-apoptotic protein that directly binds and inhibits caspases-3, 7, and 9. Here, we investigated the function of XIAP in hepatocytes in vitro using gain and loss-of-function approaches. We noted an XIAP-dependent increase in caspase activation as well as increased inflammatory markers and pro-inflammatory EV release from hepatocytes in vitro. Primary hepatocytes (PMH) from Xiap Alb.Cre and Xiap loxP mice exhibited higher cell death but surprisingly, lower expression of inflammation markers. Conditioned media from these isolated Xiap deleted PMH further decrease inflammation in bone marrow-derived macrophages. Also, interestingly, when administered an ethanol plus Fas-agonist-Jo2 model and an ethanol plus CCl4 model, these animals failed to develop an exacerbated disease phenotype in vivo. Of note, neither Xiap Alb . Cre nor Xiap AAV8.Cre mice presented with aggravated liver injury, hepatocyte apoptosis, liver steatosis, or fibrosis. Since therapeutics targeting XIAP are currently in clinical trials and caspase-induced death is very important for development of ALD, we sought to explore the potential basis of this unexpected lack of effect. We utilized scRNA-seq and spatially reconstructed hepatocyte transcriptome data from human liver tissue and observed that XIAP was significantly zonated, along with its endogenous inhibitor second mitochondria-derived activator of caspases (SMAC) in periportal region. This contrasted with pericentral zonation of other IAPs including cIAP1 and Apollon as well as caspases 3, 7, and 9. Thus providing a potential explanation for compensation of the effect of Xiap deletion by other IAPs. In conclusion, our findings implicate a potential zonallydependent role for SMAC that prevented development of a phenotype in XIAP knockout mice in ALD models. Targeting SMAC may also be important in addition to current efforts of targeting XIAP in treatment of ALD.
Collapse
Affiliation(s)
- Li He
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tejasav S. Sehrawat
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Vikas K. Verma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Amaia Navarro-Corcuera
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Guneet Sidhu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Amy Mauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xin Luo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tomohiro Katsumi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jingbiao Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Soni Shah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Juan Pablo Arab
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Sheng Cao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Hamid Kashkar
- Centre for Molecular Medicine Cologne and Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
56
|
Fang Z, Dou G, Wang L. MicroRNAs in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Int J Biol Sci 2021; 17:1851-1863. [PMID: 33994867 PMCID: PMC8120467 DOI: 10.7150/ijbs.59588] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), or, more accurately, metabolic associated fatty liver disease, accounts for a large proportion of chronic liver disorders worldwide and is closely associated with other conditions such as cardiovascular disease, obesity, and type 2 diabetes mellitus. NAFLD ranges from simple steatosis to nonalcoholic steatohepatitis (NASH) and can progress to cirrhosis and, eventually, also hepatocellular carcinoma. The morbidity and mortality associated with NAFLD are increasing rapidly year on year. Consequently, there is an urgent need to understand the etiology and pathogenesis of NAFLD and identify effective therapeutic targets. MicroRNAs (miRNAs), important epigenetic factors, have recently been proposed to participate in NAFLD pathogenesis. Here, we review the roles of miRNAs in lipid metabolism, inflammation, apoptosis, fibrosis, hepatic stellate cell activation, insulin resistance, and oxidative stress, key factors that contribute to the occurrence and progression of NAFLD. Additionally, we summarize the role of miRNA-enriched extracellular vesicles in NAFLD. These miRNAs may comprise suitable therapeutic targets for the treatment of this condition.
Collapse
Affiliation(s)
- Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Guorui Dou
- Department of Ophthalmology, Xi-Jing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
57
|
Buratta S, Shimanaka Y, Costanzi E, Ni S, Urbanelli L, Kono N, Morena F, Sagini K, Giovagnoli S, Romani R, Gargaro M, Arai H, Emiliani C. Lipotoxic stress alters the membrane lipid profile of extracellular vesicles released by Huh-7 hepatocarcinoma cells. Sci Rep 2021; 11:4613. [PMID: 33633289 PMCID: PMC7907093 DOI: 10.1038/s41598-021-84268-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are well-known mediators in intercellular communication playing pivotal roles in promoting liver inflammation and fibrosis, events associated to hepatic lipotoxicity caused by saturated free fatty acid overloading. However, despite the importance of lipids in EV membrane architecture which, in turn, affects EV biophysical and biological properties, little is known about the lipid asset of EVs released under these conditions. Here, we analyzed phospholipid profile alterations of EVs released by hepatocarcinoma Huh-7 cells under increased membrane lipid saturation induced by supplementation with saturated fatty acid palmitate or Δ9 desaturase inhibition, using oleate, a nontoxic monounsaturated fatty acid, as control. As an increase of membrane lipid saturation induces endoplasmic reticulum (ER) stress, we also analyzed phospholipid rearrangements in EVs released by Huh-7 cells treated with thapsigargin, a conventional ER stress inducer. Results demonstrate that lipotoxic and/or ER stress conditions induced rearrangements not only into cell membrane phospholipids but also into the released EVs. Thus, cell membrane saturation level and/or ER stress are crucial to determine which lipids are discarded via EVs and EV lipid cargos might be useful to discriminate hepatic lipid overloading and ER stress.
Collapse
Affiliation(s)
- Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy.
| | - Y Shimanaka
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - E Costanzi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - S Ni
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - L Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - N Kono
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - F Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - K Sagini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - S Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - R Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - M Gargaro
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - H Arai
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - C Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
58
|
Sehrawat TS, Arab JP, Liu M, Amrollahi P, Wan M, Fan J, Nakao Y, Pose E, Navarro-Corcuera A, Dasgupta D, Liao CY, He L, Mauer AS, Avitabile E, Ventura-Cots M, Bataller RA, Sanyal AJ, Chalasani NP, Heimbach JK, Watt KD, Gores GJ, Gines P, Kamath PS, Simonetto DA, Hu TY, Shah VH, Malhi H. Circulating Extracellular Vesicles Carrying Sphingolipid Cargo for the Diagnosis and Dynamic Risk Profiling of Alcoholic Hepatitis. Hepatology 2021; 73:571-585. [PMID: 32246544 PMCID: PMC7541595 DOI: 10.1002/hep.31256] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/27/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Alcoholic hepatitis (AH) is diagnosed by clinical criteria, although several objective scores facilitate risk stratification. Extracellular vesicles (EVs) have emerged as biomarkers for many diseases and are also implicated in the pathogenesis of AH. Therefore, we investigated whether plasma EV concentration and sphingolipid cargo could serve as diagnostic biomarkers for AH and inform prognosis to permit dynamic risk profiling of AH subjects. APPROACH AND RESULTS EVs were isolated and quantified from plasma samples from healthy controls, heavy drinkers, and subjects with end-stage liver disease (ESLD) attributed to cholestatic liver diseases and nonalcoholic steatohepatitis, decompensated alcohol-associated cirrhosis (AC), and AH. Sphingolipids were quantified by tandem mass spectroscopy. The median plasma EV concentration was significantly higher in AH subjects (5.38 × 1011 /mL) compared to healthy controls (4.38 × 1010 /mL; P < 0.0001), heavy drinkers (1.28 × 1011 /mL; P < 0.0001), ESLD (5.35 × 1010 /mL; P < 0.0001), and decompensated AC (9.2 × 1010 /mL; P < 0.0001) disease controls. Among AH subjects, EV concentration correlated with Model for End-Stage Liver Disease score. When EV counts were dichotomized at the median, survival probability for AH subjects at 90 days was 63.0% in the high-EV group and 90.0% in the low-EV group (log-rank P value = 0.015). Interestingly, EV sphingolipid cargo was significantly enriched in AH when compared to healthy controls, heavy drinkers, ESLD, and decompensated AC (P = 0.0001). Multiple sphingolipids demonstrated good diagnostic and prognostic performance as biomarkers for AH. CONCLUSIONS Circulating EV concentration and sphingolipid cargo signature can be used in the diagnosis and differentiation of AH from heavy drinkers, decompensated AC, and other etiologies of ESLD and predict 90-day survival permitting dynamic risk profiling.
Collapse
Affiliation(s)
| | - Juan P Arab
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN.,Departamento de GastroenterologiaEscuela de MedicinaPontificia Universidad Catolica de ChileSantiagoChile
| | - Mengfei Liu
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | - Pouya Amrollahi
- Virginia G. Piper Biodesign Center for Personalized DiagnosticsThe Biodesign InstituteArizona State UniversityTempeAZ.,School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ
| | - Meihua Wan
- Virginia G. Piper Biodesign Center for Personalized DiagnosticsThe Biodesign InstituteArizona State UniversityTempeAZ.,School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ.,Department of Integrated Traditional Chinese and Western MedicineWest China Hospital of Sichuan UniversityChengduChina
| | - Jia Fan
- Virginia G. Piper Biodesign Center for Personalized DiagnosticsThe Biodesign InstituteArizona State UniversityTempeAZ.,School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ
| | - Yasuhiko Nakao
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN.,Nagasaki University HospitalNagasakiJapan
| | - Elisa Pose
- Liver UnitHospital Clínic de BarcelonaSchool of Medicine and Health SciencesUniversity of BarcelonaBarcelonaSpain.,Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS)BarcelonaSpain.,Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (CIBEReHD)BarcelonaSpain
| | | | | | - Chieh-Yu Liao
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | - Li He
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN.,Division of GastroenterologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Amy S Mauer
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | - Emma Avitabile
- Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Meritxell Ventura-Cots
- Division of Gastroenterology, Hepatology and NutritionUniversity of Pittsburgh Medical CenterPittsburghPA
| | - Ramon A Bataller
- Division of Gastroenterology, Hepatology and NutritionUniversity of Pittsburgh Medical CenterPittsburghPA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and NutritionVirginia Commonwealth UniversityRichmondVA
| | - Naga P Chalasani
- Division of Gastroenterology and HepatologyIndiana UniversityIndianapolisIN
| | | | - Kymberly D Watt
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | - Gregory J Gores
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | - Pere Gines
- Liver UnitHospital Clínic de BarcelonaSchool of Medicine and Health SciencesUniversity of BarcelonaBarcelonaSpain.,Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS)BarcelonaSpain.,Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (CIBEReHD)BarcelonaSpain
| | - Patrick S Kamath
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | | | - Tony Y Hu
- Virginia G. Piper Biodesign Center for Personalized DiagnosticsThe Biodesign InstituteArizona State UniversityTempeAZ.,School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ
| | - Vijay H Shah
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| | - Harmeet Malhi
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMN
| |
Collapse
|
59
|
Kjær MB, George J, Kazankov K, Grønbæk H. Current perspectives on the pathophysiology of metabolic associated fatty liver disease: are macrophages a viable target for therapy? Expert Rev Gastroenterol Hepatol 2021; 15:51-64. [PMID: 32878486 DOI: 10.1080/17474124.2020.1817740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Metabolic associated fatty liver disease (MAFLD) is a new nomenclature for fatty liver replacing nonalcoholic fatty liver disease (NAFLD). MAFLD has emerged as the leading cause of liver-related morbidity and mortality with increasing incidence due to its close association with the global epidemic of obesity and type 2 diabetes mellitus. Macrophages play a key role in MAFLD development and progression of steatohepatitis and fibrosis. Therefore, targeting macrophages may be a new therapeutic approach for MAFLD and MAFLD with steatohepatitis. AREAS COVERED We provide a comprehensive review of the significant role of macrophages in MAFLD. Further, we evaluate the current status of lifestyle interventions and pharmacological treatments with a focus on effects mediated through direct or indirect targeting of macrophages. EXPERT OPINION Targeting macrophages holds promise as a treatment option for the management of MAFLD and steatohepatitis. Improved stratification of patients according to MAFLD phenotype would contribute to more adequate design enhancing the yield of clinical trials ultimately leading to personalized medicine for patients with MAFLD. Furthermore, reflecting the multifactorial pathogenesis of MAFLD, combination therapies based on the various pathophysiological driver events including as pertinent to this review, macrophage recruitment, polarization and action, present an intriguing target for future investigation.
Collapse
Affiliation(s)
- Mikkel Breinholt Kjær
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus, Denmark
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney , Sydney, Australia
| | - Konstantin Kazankov
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus, Denmark
| | - Henning Grønbæk
- Department of Hepatology and Gastroenterology, Aarhus University Hospital , Aarhus, Denmark
| |
Collapse
|
60
|
van Meteren N, Lagadic-Gossmann D, Podechard N, Gobart D, Gallais I, Chevanne M, Collin A, Burel A, Dupont A, Rault L, Chevance S, Gauffre F, Le Ferrec E, Sergent O. Extracellular vesicles released by polycyclic aromatic hydrocarbons-treated hepatocytes trigger oxidative stress in recipient hepatocytes by delivering iron. Free Radic Biol Med 2020; 160:246-262. [PMID: 32791186 DOI: 10.1016/j.freeradbiomed.2020.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/14/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
Abstract
A growing body of evidences indicate the major role of extracellular vesicles (EVs) as players of cell communication in the pathogenesis of liver diseases. EVs are membrane-enclosed vesicles released by cells into the extracellular environment. Oxidative stress is also a key component of liver disease pathogenesis, but no role for hepatocyte-derived EVs has yet been described in the development of this process. Recently, some polycyclic aromatic hydrocarbons (PAHs), widespread environmental contaminants, were demonstrated to induce EV release from hepatocytes. They are also well-known to trigger oxidative stress leading to cell death. Therefore, the aim of this work was to investigate the involvement of EVs derived from PAHs-treated hepatocytes (PAH-EVs) in possible oxidative damages of healthy recipient hepatocytes, using both WIF-B9 and primary rat hepatocytes. We first showed that the release of EVs from PAHs -treated hepatocytes depended on oxidative stress. PAH-EVs were enriched in proteins related to oxidative stress such as NADPH oxidase and ferritin. They were also demonstrated to contain more iron. PAH-EVs could then induce oxidative stress in recipient hepatocytes, thereby leading to apoptosis. Mitochondria and lysosomes of recipient hepatocytes exhibited significant structural alterations. All those damages were dependent on internalization of EVs that reached lysosomes with their cargoes. Lysosomes thus appeared as critical organelles for EVs to induce apoptosis. In addition, pro-oxidant components of PAH-EVs, e.g. NADPH oxidase and iron, were revealed to be necessary for this cell death.
Collapse
Affiliation(s)
- Nettie van Meteren
- 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
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Dimitri Gobart
- 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
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Aurore Collin
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Agnès Burel
- Univ Rennes, Biosit, UMS 3480, US_S 018, F-35000, Rennes, France
| | - Aurélien Dupont
- Univ Rennes, Biosit, UMS 3480, US_S 018, F-35000, Rennes, France
| | | | - Soizic Chevance
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Fabienne Gauffre
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Eric Le Ferrec
- 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.
| |
Collapse
|
61
|
Dorairaj V, Sulaiman SA, Abu N, Abdul Murad NA. Extracellular Vesicles in the Development of the Non-Alcoholic Fatty Liver Disease: An Update. Biomolecules 2020; 10:biom10111494. [PMID: 33143043 PMCID: PMC7693409 DOI: 10.3390/biom10111494] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a broad spectrum of liver damage disease from a simple fatty liver (steatosis) to more severe liver conditions such as non-alcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. Extracellular vesicles (EVs) are a heterogeneous group of small membrane vesicles released by various cells in normal or diseased conditions. The EVs carry bioactive components in their cargos and can mediate the metabolic changes in recipient cells. In the context of NAFLD, EVs derived from adipocytes are implicated in the development of whole-body insulin resistance (IR), the hepatic IR, and fatty liver (steatosis). Excessive fatty acid accumulation is toxic to the hepatocytes, and this lipotoxicity can induce the release of EVs (hepatocyte-EVs), which can mediate the progression of fibrosis via the activation of nearby macrophages and hepatic stellate cells (HSCs). In this review, we summarized the recent findings of adipocyte- and hepatocyte-EVs on NAFLD disease development and progression. We also discussed previous studies on mesenchymal stem cell (MSC) EVs that have garnered attention due to their effects on preventing liver fibrosis and increasing liver regeneration and proliferation.
Collapse
|
62
|
Gwag T, Reddy Mooli RG, Li D, Lee S, Lee EY, Wang S. Macrophage-derived thrombospondin 1 promotes obesity-associated non-alcoholic fatty liver disease. JHEP Rep 2020; 3:100193. [PMID: 33294831 PMCID: PMC7689554 DOI: 10.1016/j.jhepr.2020.100193] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background & Aims Thrombospondin 1 (TSP1) is a multifunctional matricellular protein. We previously showed that TSP1 has an important role in obesity-associated metabolic complications, including inflammation, insulin resistance, cardiovascular, and renal disease. However, its contribution to obesity-associated non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD or NASH) remains largely unknown; thus, we aimed to determine its role. Methods High-fat diet or AMLN (amylin liver NASH) diet-induced obese and insulin-resistant NAFLD/NASH mouse models were utilised, in addition to tissue-specific Tsp1-knockout mice, to determine the contribution of different cellular sources of obesity-induced TSP1 to NAFLD/NASH development. Results Liver TSP1 levels were increased in experimental obese and insulin-resistant NAFLD/NASH mouse models as well as in obese patients with NASH. Moreover, TSP1 deletion in adipocytes did not protect mice from diet-induced NAFLD/NASH. However, myeloid/macrophage-specific TSP1 deletion protected mice against obesity-associated liver injury, accompanied by reduced liver inflammation and fibrosis. Importantly, this protection was independent of the levels of obesity and hepatic steatosis. Mechanistically, through an autocrine effect, macrophage-derived TSP1 suppressed Smpdl3b expression in liver, which amplified liver proinflammatory signalling (Toll-like receptor 4 signal pathway) and promoted NAFLD progression. Conclusions Macrophage-derived TSP1 is a significant contributor to obesity-associated NAFLD/NASH development and progression and could serve as a therapeutic target for this disease. Lay summary Obesity-associated non-alcoholic fatty liver disease is a most common chronic liver disease in the Western world and can progress to liver cirrhosis and cancer. No treatment is currently available for this disease. The present study reveals an important factor (macrophage-derived TSP1) that drives macrophage activation and non-alcoholic fatty liver disease development and progression and that could serve as a therapeutic target for non-alcoholic fatty liver disease/steatohepatitis.
Collapse
Key Words
- ALT, alanine aminotransferase
- AMLN, amylin liver NASH
- ASMase, acid sphingomyelinase
- AST, aspartate aminotransferase
- BMDM, bone marrow-derived macrophage
- DEG, differentially expressed gene
- EC, endothelial cell
- ECM, extracellular matrix
- GPI, glycosylphosphatidylinositol
- HFD, high-fat diet
- HSC, hepatic stellate cell
- IL-, interleukin-
- KC, Kupffer cell
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LFD, low-fat diet
- LPS, lipopolysaccharide
- MDM, monocyte-derived macrophage
- MP, mononuclear phagocyte
- Macrophage
- NAFLD
- NAFLD, non-alcoholic fatty liver disease
- NAS, NAFLD activity score
- NASH
- NASH, non-alcoholic steatohepatitis
- NF-κB, nuclear factor-κB
- Obesity
- SMPDL3B
- SMPDL3B, sphingomyelin phosphodiesterase acid-like 3B
- SREBP1c, sterol regulatory element-binding protein-1 c
- TGF, transforming growth factor
- TLR, Toll-like receptor
- TNF, tumour necrosis factor
- TSP1
- TSP1, thrombospondin 1
- Th, T helper type
- Tsp1fl/fl, TSP1 floxed mice
- Tsp1Δadipo, adipocyte-specific TSP1-knockout mice
- Tsp1Δmɸ, macrophage-specific TSP1-knockout mice
- qPCR, quantitative PCR
- scRNA-seq, single-cell RNA sequencing
- α-SMA, smooth muscle actin
Collapse
Affiliation(s)
- Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Raja Gopal Reddy Mooli
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Dong Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Sangderk Lee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Eun Y Lee
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
63
|
Dasgupta D, Nakao Y, Mauer AS, Thompson JM, Sehrawat TS, Liao CY, Krishnan A, Lucien F, Guo Q, Liu M, Xue F, Fukushima M, Katsumi T, Bansal A, Pandey MK, Maiers JL, DeGrado T, Ibrahim SH, Revzin A, Pavelko KD, Barry MA, Kaufman RJ, Malhi H. IRE1A Stimulates Hepatocyte-Derived Extracellular Vesicles That Promote Inflammation in Mice With Steatohepatitis. Gastroenterology 2020; 159:1487-1503.e17. [PMID: 32574624 PMCID: PMC7666601 DOI: 10.1053/j.gastro.2020.06.031] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 05/14/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Endoplasmic reticulum to nucleus signaling 1 (ERN1, also called IRE1A) is a sensor of the unfolded protein response that is activated in the livers of patients with nonalcoholic steatohepatitis (NASH). Hepatocytes release ceramide-enriched inflammatory extracellular vesicles (EVs) after activation of IRE1A. We studied the effects of inhibiting IRE1A on release of inflammatory EVs in mice with diet-induced steatohepatitis. METHODS C57BL/6J mice and mice with hepatocyte-specific disruption of Ire1a (IRE1αΔhep) were fed a diet high in fat, fructose, and cholesterol to induce development of steatohepatitis or a standard chow diet (controls). Some mice were given intraperitoneal injections of the IRE1A inhibitor 4μ8C. Mouse liver and primary hepatocytes were transduced with adenovirus or adeno-associated virus that expressed IRE1A. Livers were collected from mice and analyzed by quantitative polymerase chain reaction and chromatin immunoprecipitation assays; plasma samples were analyzed by enzyme-linked immunosorbent assay. EVs were derived from hepatocytes and injected intravenously into mice. Plasma EVs were characterized by nanoparticle-tracking analysis, electron microscopy, immunoblots, and nanoscale flow cytometry; we used a membrane-tagged reporter mouse to detect hepatocyte-derived EVs. Plasma and liver tissues from patients with NASH and without NASH (controls) were analyzed for EV concentration and by RNAscope and gene expression analyses. RESULTS Disruption of Ire1a in hepatocytes or inhibition of IRE1A reduced the release of EVs and liver injury, inflammation, and accumulation of macrophages in mice on the diet high in fat, fructose, and cholesterol. Activation of IRE1A, in the livers of mice, stimulated release of hepatocyte-derived EVs, and also from cultured primary hepatocytes. Mice given intravenous injections of IRE1A-stimulated, hepatocyte-derived EVs accumulated monocyte-derived macrophages in the liver. IRE1A-stimulated EVs were enriched in ceramides. Chromatin immunoprecipitation showed that IRE1A activated X-box binding protein 1 (XBP1) to increase transcription of serine palmitoyltransferase genes, which encode the rate-limiting enzyme for ceramide biosynthesis. Administration of a pharmacologic inhibitor of serine palmitoyltransferase to mice reduced the release of EVs. Levels of XBP1 and serine palmitoyltransferase were increased in liver tissues, and numbers of EVs were increased in plasma, from patients with NASH compared with control samples and correlated with the histologic features of inflammation. CONCLUSIONS In mouse hepatocytes, activated IRE1A promotes transcription of serine palmitoyltransferase genes via XBP1, resulting in ceramide biosynthesis and release of EVs. The EVs recruit monocyte-derived macrophages to the liver, resulting in inflammation and injury in mice with diet-induced steatohepatitis. Levels of XBP1, serine palmitoyltransferase, and EVs are all increased in liver tissues from patients with NASH. Strategies to block this pathway might be developed to reduce liver inflammation in patients with NASH.
Collapse
Affiliation(s)
- Debanjali Dasgupta
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Yasuhiko Nakao
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905,Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Amy S Mauer
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Jill M Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905
| | - Tejasav S Sehrawat
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Chieh-Yu Liao
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Anuradha Krishnan
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | | | - Qianqian Guo
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Mengfei Liu
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Fei Xue
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Masanori Fukushima
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905,Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Tomohiro Katsumi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905,2-2-2 Iidanishi Yamagata city, Yamagata, Japan 990-9585 Yamagata University Faculty of Medicine, Department of Gastroenterology
| | - Aditya Bansal
- Department of Radiology, Mayo Clinic, Rochester, MN 55905
| | | | - Jessica L Maiers
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | | | - Samar H Ibrahim
- Division of Pediatric Gastroenterology, Mayo Clinic, Rochester, MN 55905
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905
| | | | - Michael A Barry
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905
| | - Randal J Kaufman
- Center for Neuroscience, Aging, and Stem Cell Research, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
64
|
Yang D, Liu J. Targeting extracellular vesicles-mediated hepatic inflammation as a therapeutic strategy in liver diseases. Liver Int 2020; 40:2064-2073. [PMID: 32593200 DOI: 10.1111/liv.14579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/18/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Extracellular Vesicles (EVs) are nano- to micro-sized membranous vesicles that can be produced by normal and diseased cells. As carriers of biologically active molecules including proteins, lipids and nucleic acids, EVs mediate cell-to-cell communication and execute diverse functions by delivering their cargoes to specific cell types. Hepatic inflammation caused by virus infection, autoimmunity and malignancy is a common driver of progressive liver fibrosis and permanent liver damage. Emerging evidence has shown that EVs-mediated inflammation as critical player in the progression of liver diseases since they shuttle within the liver as well as between other tissues with inflammatory signals. Therefore, targeting inflammatory EVs could represent a potential therapeutic strategy in liver diseases. Moreover, EVs are emerging as a promising tool for intracellular delivery of therapeutic nucleic acid. In this review, we will discuss not only recent advances on the role of EVs in mediating hepatic inflammation and present strategies for targeted therapy on the context of liver diseases but also the challenging questions that need to be answered in the field.
Collapse
Affiliation(s)
- Dakai Yang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Jing Liu
- Microbiology and Immunity Department, Shanghai University of Medicine & Health Sciences, Shanghai, People's Republic of China
| |
Collapse
|
65
|
Guitton J, Bandet CL, Mariko ML, Tan-Chen S, Bourron O, Benomar Y, Hajduch E, Le Stunff H. Sphingosine-1-Phosphate Metabolism in the Regulation of Obesity/Type 2 Diabetes. Cells 2020; 9:E1682. [PMID: 32668665 PMCID: PMC7407406 DOI: 10.3390/cells9071682] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is a pathophysiological condition where excess free fatty acids (FFA) target and promote the dysfunctioning of insulin sensitive tissues and of pancreatic β cells. This leads to the dysregulation of glucose homeostasis, which culminates in the onset of type 2 diabetes (T2D). FFA, which accumulate in these tissues, are metabolized as lipid derivatives such as ceramide, and the ectopic accumulation of the latter has been shown to lead to lipotoxicity. Ceramide is an active lipid that inhibits the insulin signaling pathway as well as inducing pancreatic β cell death. In mammals, ceramide is a key lipid intermediate for sphingolipid metabolism as is sphingosine-1-phosphate (S1P). S1P levels have also been associated with the development of obesity and T2D. In this review, the current knowledge on S1P metabolism in regulating insulin signaling in pancreatic β cell fate and in the regulation of feeding by the hypothalamus in the context of obesity and T2D is summarized. It demonstrates that S1P can display opposite effects on insulin sensitive tissues and pancreatic β cells, which depends on its origin or its degradation pathway.
Collapse
Affiliation(s)
- Jeanne Guitton
- Institut des Neurosciences Paris-Saclay, Université Paris Saclay, CNRS UMR 9197, F-91190 Orsay, France; (J.G.); (M.L.M.); (Y.B.)
| | - Cecile L. Bandet
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (C.L.B.); (S.T.-C.); (O.B.); (E.H.)
- Institut Hospitalo-Universitaire ICAN, F-75013 Paris, France
| | - Mohamed L. Mariko
- Institut des Neurosciences Paris-Saclay, Université Paris Saclay, CNRS UMR 9197, F-91190 Orsay, France; (J.G.); (M.L.M.); (Y.B.)
| | - Sophie Tan-Chen
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (C.L.B.); (S.T.-C.); (O.B.); (E.H.)
- Institut Hospitalo-Universitaire ICAN, F-75013 Paris, France
| | - Olivier Bourron
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (C.L.B.); (S.T.-C.); (O.B.); (E.H.)
- Institut Hospitalo-Universitaire ICAN, F-75013 Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Diabétologie et Maladies métaboliques, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Yacir Benomar
- Institut des Neurosciences Paris-Saclay, Université Paris Saclay, CNRS UMR 9197, F-91190 Orsay, France; (J.G.); (M.L.M.); (Y.B.)
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (C.L.B.); (S.T.-C.); (O.B.); (E.H.)
- Institut Hospitalo-Universitaire ICAN, F-75013 Paris, France
| | - Hervé Le Stunff
- Institut des Neurosciences Paris-Saclay, Université Paris Saclay, CNRS UMR 9197, F-91190 Orsay, France; (J.G.); (M.L.M.); (Y.B.)
| |
Collapse
|
66
|
Ministrini S, Montecucco F, Sahebkar A, Carbone F. Macrophages in the pathophysiology of NAFLD: The role of sex differences. Eur J Clin Invest 2020; 50:e13236. [PMID: 32294235 DOI: 10.1111/eci.13236] [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] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 03/14/2020] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a multifactorial pathological condition, which recognizes a certain sexual dimorphism. Experimental and clinical studies provided evidence for a critical role of macrophages in NAFLD development and progression. Especially, liver-resident macrophages (also known as Kupffer cells) are likely the common final pathway of several pro-steatosic signals. A huge amount of danger-associated molecular patterns recognized by Kupffer cells is provided within the liver by lipid and glucose toxicity. Other pro-inflammatory signals come from surrounding tissues into the portal vein, directly to the liver: they come from dysfunctional adipocytes, adipose tissue macrophages and gut dysbiosis. These complex crosstalks are differently represented across sexes, as sexual hormones control many of these processes. Sexual dimorphism then modulates metabolic and inflammatory cascades driving the liver from a simple steatosis to NAFLD and beyond. Here, metabolic and inflammatory mechanisms underlying NALFD pathophysiology will be updated. A special attention will be paid to describe sex-related differences that could provide insights for patient stratification and more tailored therapeutic approaches.
Collapse
Affiliation(s)
- Stefano Ministrini
- Internal Medicine Department, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- First Clinic of Internal Medicine Department of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
67
|
Shaping of Innate Immune Response by Fatty Acid Metabolite Palmitate. Cells 2019; 8:cells8121633. [PMID: 31847240 PMCID: PMC6952933 DOI: 10.3390/cells8121633] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Innate immune cells monitor invading pathogens and pose the first-line inflammatory response to coordinate with adaptive immunity for infection removal. Innate immunity also plays pivotal roles in injury-induced tissue remodeling and the maintenance of tissue homeostasis in physiological and pathological conditions. Lipid metabolites are emerging as the key players in the regulation of innate immune responses, and recent work has highlighted the importance of the lipid metabolite palmitate as an essential component in this regulation. Palmitate modulates innate immunity not only by regulating the activation of pattern recognition receptors in local innate immune cells, but also via coordinating immunological activity in inflammatory tissues. Moreover, protein palmitoylation controls various cellular physiological processes. Herein, we review the updated evidence that palmitate catabolism contributes to innate immune cell-mediated inflammatory processes that result in immunometabolic disorders.
Collapse
|
68
|
Abstract
Extracellular vesicles (EVs) are membrane-defined nanoparticles released by most cell types. The EVs released by cells may differ quantitatively and qualitatively from physiological states to disease states. There are several unique properties of EVs, including their proteins, lipids and nucleic acid cargoes, stability in circulation, and presence in biofluids, which make them a critical vector for cell-to-cell communication and impart utility as a biomarker. EVs may also serve as a vehicle for selective cargo secretion. Similarly, EV cargo may be selectively manipulated for targeted therapeutic delivery. In this review an overview is provided on the EV classification, biogenesis, and secretion pathways, which are conserved across cell types. Next, cargo characterization and effector cell responses are discussed in the context of nonalcoholic steatohepatitis, alcoholic hepatitis, and acetaminophen-induced liver injury. The review also discusses the potential biomarker and therapeutic uses of circulating EVs.
Collapse
Affiliation(s)
- Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
69
|
Balaphas A, Meyer J, Sadoul R, Morel P, Gonelle-Gispert C, Bühler LH. Extracellular vesicles: Future diagnostic and therapeutic tools for liver disease and regeneration. Liver Int 2019; 39:1801-1817. [PMID: 31286675 DOI: 10.1111/liv.14189] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/06/2019] [Accepted: 07/01/2019] [Indexed: 02/13/2023]
Abstract
Extracellular vesicles are membrane fragments that can be produced by all cell types. Interactions between extracellular vesicles and various liver cells constitute an emerging field in hepatology and recent evidences have established a role for extracellular vesicles in various liver diseases and physiological processes. Extracellular vesicles originating from liver cells are implicated in intercellular communication and fluctuations of specific circulating extracellular vesicles could constitute new diagnostic tools. In contrast, extracellular vesicles derived from progenitor cells interact with hepatocytes or non-parenchymal cells, thereby protecting the liver from various injuries and promoting liver regeneration. Our review focuses on recent developments investigating the role of various types of extracellular vesicles in acute and chronic liver diseases as well as their potential use as biomarkers and therapeutic tools.
Collapse
Affiliation(s)
- Alexandre Balaphas
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Jeremy Meyer
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Rémy Sadoul
- Université Grenoble Alpes, Institut des Neurosciences, Grenoble, France
| | - Philippe Morel
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Leo Hans Bühler
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
70
|
Korbecki J, Bajdak-Rusinek K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm Res 2019; 68:915-932. [PMID: 31363792 PMCID: PMC6813288 DOI: 10.1007/s00011-019-01273-5] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Palmitic acid is a saturated fatty acid whose blood concentration is elevated in obese patients. This causes inflammatory responses, where toll-like receptors (TLR), TLR2 and TLR4, play an important role. Nevertheless, palmitic acid is not only a TLR agonist. In the cell, this fatty acid is converted into phospholipids, diacylglycerol and ceramides. They trigger the activation of various signaling pathways that are common for LPS-mediated TLR4 activation. In particular, metabolic products of palmitic acid affect the activation of various PKCs, ER stress and cause an increase in ROS generation. Thanks to this, palmitic acid also strengthens the TLR4-induced signaling. In this review, we discuss the mechanisms of inflammatory response induced by palmitic acid. In particular, we focus on describing its effect on ER stress and IRE1α, and the mechanisms of NF-κB activation. We also present the mechanisms of inflammasome NLRP3 activation and the effect of palmitic acid on enhanced inflammatory response by increasing the expression of FABP4/aP2. Finally, we focus on the consequences of inflammatory responses, in particular, the effect of TNF-α, IL-1β and IL-6 on insulin resistance. Due to the high importance of macrophages and the production of proinflammatory cytokines by them, this work mainly focuses on these cells.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Molecular Biology, School of Medicine in Katowice, Medical University of Silesia, Medyków 18 St., 40-752, Katowice, Poland.
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, School of Medicine in Katowice, Medical University of Silesia, Medyków 18 St., 40-752, Katowice, Poland
| |
Collapse
|
71
|
Weigert A, Olesch C, Brüne B. Sphingosine-1-Phosphate and Macrophage Biology-How the Sphinx Tames the Big Eater. Front Immunol 2019; 10:1706. [PMID: 31379883 PMCID: PMC6658986 DOI: 10.3389/fimmu.2019.01706] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022] Open
Abstract
The sphingolipid sphingosine-1-phosphate (S1P) is produced by sphingosine kinases to either signal through intracellular targets or to activate a family of specific G-protein-coupled receptors (S1PR). S1P levels are usually low in peripheral tissues compared to the vasculature, forming a gradient that mediates lymphocyte trafficking. However, S1P levels rise during inflammation in peripheral tissues, thereby affecting resident or recruited immune cells, including macrophages. As macrophages orchestrate initiation and resolution of inflammation, the sphingosine kinase/S1P/S1P-receptor axis emerges as an important determinant of macrophage function in the pathogenesis of inflammatory diseases such as cancer, atherosclerosis, and infection. In this review, we therefore summarize the current knowledge how S1P affects macrophage biology.
Collapse
Affiliation(s)
- Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Catherine Olesch
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
72
|
Li J, Liu H, Mauer AS, Lucien F, Raiter A, Bandla H, Mounajjed T, Yin Z, Glaser KJ, Yin M, Malhi H. Characterization of Cellular Sources and Circulating Levels of Extracellular Vesicles in a Dietary Murine Model of Nonalcoholic Steatohepatitis. Hepatol Commun 2019; 3:1235-1249. [PMID: 31497744 PMCID: PMC6719742 DOI: 10.1002/hep4.1404] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/24/2019] [Indexed: 12/18/2022] Open
Abstract
Circulating extracellular vesicles (EVs) are a novel and emerging biomarker for nonalcoholic steatohepatitis (NASH). It has been demonstrated that total circulating EVs and hepatocyte‐derived EVs are elevated in male mice with diet‐induced NASH. How hepatocyte‐derived EVs change over time and other cellular sources of EVs in NASH have not been determined. Our objective was to define the quantitative evolution of hepatocyte‐derived, macrophage‐derived, neutrophil‐derived, and platelet‐derived EVs in male and female mice with dietary NASH. Fluorescently labeled antibodies and a nanoscale flow cytometer were used to detect plasma levels of EVs. Asialoglycoprotein receptor 1 (ASGR1) and cytochrome P450 family 2 subfamily E member 1 (CYP2E1) are markers of hepatocyte‐derived EVs; galectin 3 is a marker of macrophage‐derived EVs; common epitope on lymphocyte antigen 6 complex, locus G/C1 (Ly‐6G and Ly‐6C) is a marker of neutrophil‐derived EVs; and clusters of differentiation 61 (CD61) is a marker of platelet‐derived EVs. Nonalcoholic fatty liver disease activity score (NAS) was calculated using hematoxylin and eosin‐stained liver sections, and magnetic resonance imaging (MRI) was used for measurement of the fat fraction and elastography. Hepatocyte‐derived EVs increased in both male and female mice at 12 and 10 weeks of feeding, respectively, and remained elevated at 24 weeks in both male and female mice and at 48 weeks in male mice and 36 weeks in female mice. Macrophage‐ and neutrophil‐derived EVs were significantly elevated at 24 weeks of dietary feeding concomitant with the histologic presence of inflammatory foci in the liver. In fat‐, fructose‐, and cholesterol‐ (FFC) fed male mice, platelet‐derived EVs were elevated at 12, 24, and 48 weeks, whereas in female mice, platelet derived EVs were significantly elevated at 24 weeks. Hepatocyte‐, macrophage‐ and neutrophil‐derived EVs correlated well with the histologic NAS. Conclusion: Circulating cell‐type‐specific EVs may be a novel biomarker for NASH diagnosis and longitudinal follow up.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Radiology Mayo Clinic Rochester MN
| | - Huimin Liu
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester MN.,Center of Integrative Medicine Beijing Ditan Hospital Capital Medical University Beijing China
| | - Amy S Mauer
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester MN
| | | | - Abagail Raiter
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester MN
| | | | | | - Ziying Yin
- Department of Radiology Mayo Clinic Rochester MN
| | | | - Meng Yin
- Department of Radiology Mayo Clinic Rochester MN
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology Mayo Clinic Rochester MN
| |
Collapse
|