101
|
Metabolic Nano-Machines: Extracellular Vesicles Containing Active Enzymes and Their Contribution to Liver Diseases. CURRENT PATHOBIOLOGY REPORTS 2019. [DOI: 10.1007/s40139-019-00197-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
102
|
Gloor Y, Schvartz D, F Samer C. Old problem, new solutions: biomarker discovery for acetaminophen liver toxicity. Expert Opin Drug Metab Toxicol 2019; 15:659-669. [PMID: 31293190 DOI: 10.1080/17425255.2019.1642323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Although the hepatotoxicity of acetaminophen is a well-known problem, the search for reliable biomarker of toxicity is still a current issue as clinical tools are missing to assess patients intoxicated following chronic use, sequential ingestion, use of modified release formulations or in case of delayed arrival to hospital. The need for new specific and robust biomarkers for acetaminophen toxicity has prompted many studies exploring the use of blood levels of acetaminophen derivatives, mitochondrial damage markers, liver cell apoptosis and/or necrosis markers and circulating microRNAs. Areas covered: In this review, we present a concise overview of the most promising biomarkers currently under evaluation including descriptions of their properties with respect to exposure type, APAP specificity, and potential clinical application. In addition, we illustrate the power of new technologies for biomarker research and describe their current application to the field of acetaminophen-induced hepatotoxicity. Expert opinion: Recently the use of extracellular vesicles isolation in combination with omics techniques has opened a new perspective to the field of biomarker research. However, the potential of those new technologies for the prediction and monitoring of hepatic diseases and acetaminophen toxicity has not yet been fully taken into consideration.
Collapse
Affiliation(s)
- Yvonne Gloor
- a Division of clinical pharmacology and toxicology, Geneva University Hospital , Geneva , Switzerland.,b Laboratory of clinical pharmacology, Faculty of Medicine, University of Geneva , Geneva , Switzerland
| | - Domitille Schvartz
- c Department of internal medicine specialties, Faculty of Medicine, University of Geneva , Geneva , Switzerland
| | - Caroline F Samer
- a Division of clinical pharmacology and toxicology, Geneva University Hospital , Geneva , Switzerland.,b Laboratory of clinical pharmacology, Faculty of Medicine, University of Geneva , Geneva , Switzerland
| |
Collapse
|
103
|
McDaniel K, Wu N, Zhou T, Huang L, Sato K, Venter J, Ceci L, Chen D, Ramos-Lorenzo S, Invernizzi P, Bernuzzi F, Wu C, Francis H, Glaser S, Alpini G, Meng F. Amelioration of Ductular Reaction by Stem Cell Derived Extracellular Vesicles in MDR2 Knockout Mice via Lethal-7 microRNA. Hepatology 2019; 69:2562-2578. [PMID: 30723922 PMCID: PMC7015419 DOI: 10.1002/hep.30542] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
Abstract
Cholangiopathies are diseases that affect cholangiocytes, the cells lining the biliary tract. Liver stem cells (LSCs) are able to differentiate into all cells of the liver and possibly influence the surrounding liver tissue by secretion of signaling molecules. One way in which cells can interact is through secretion of extracellular vesicles (EVs), which are small membrane-bound vesicles that contain proteins, microRNAs (miRNAs), and cytokines. We evaluated the contents of liver stem cell-derived EVs (LSCEVs), compared their miRNA contents to those of EVs isolated from hepatocytes, and evaluated the downstream targets of these miRNAs. We finally evaluated the crosstalk among LSCs, cholangiocytes, and human hepatic stellate cells (HSCs). We showed that LSCEVs were able to reduce ductular reaction and biliary fibrosis in multidrug resistance protein 2 (MDR2)-/- mice. Additionally, we showed that cholangiocyte growth was reduced and HSCs were deactivated in LSCEV-treated mice. Evaluation of LSCEV contents compared with EVs derived from hepatocytes showed a large increase in the miRNA, lethal-7 (let-7). Further evaluation of let-7 in MDR2-/- mice and human primary sclerosing cholangitis samples showed reduced levels of let-7 compared with controls. In liver tissues and isolated cholangiocytes, downstream targets of let-7 (identified by ingenuity pathway analysis), Lin28a (Lin28 homolog A), Lin28b (Lin28 homolog B), IL-13 (interleukin 13), NR1H4 (nuclear receptor subfamily 1 group H member 4) and NF-κB (nuclear factor kappa B), are elevated in MDR2-/- mice, but treatment with LSCEVs reduced levels of these mediators of ductular reaction and biliary fibrosis through the inhibition of NF-κB and IL-13 signaling pathways. Evaluation of crosstalk using cholangiocyte supernatants from LSCEV-treated cells on cultured HSCs showed that HSCs had reduced levels of fibrosis and increased senescence. Conclusion: Our studies indicate that LSCEVs could be a possible treatment for cholangiopathies or could be used for target validation for future therapies.
Collapse
Affiliation(s)
- Kelly McDaniel
- Research Department, Central Texas Veterans Health Care System, Temple, TX
- Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX
| | - Nan Wu
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Tianhao Zhou
- Research Department, Central Texas Veterans Health Care System, Temple, TX
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Li Huang
- Department of Pancreatobiliary Surgery and Center for Translational Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Keisaku Sato
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Julie Venter
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Ludovica Ceci
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Demeng Chen
- Department of Pancreatobiliary Surgery and Center for Translational Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sugeily Ramos-Lorenzo
- Research Department, Central Texas Veterans Health Care System, Temple, TX
- Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX
| | - Pietro Invernizzi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Francesca Bernuzzi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX
| | - Heather Francis
- Research Department, Central Texas Veterans Health Care System, Temple, TX
- Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Shannon Glaser
- Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Gianfranco Alpini
- Research Department, Central Texas Veterans Health Care System, Temple, TX
- Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX
- Department of Medical Physiology, Texas A&M University Health Science Center College of Medicine, Temple, TX
| | - Fanyin Meng
- Research Department, Central Texas Veterans Health Care System, Temple, TX
- Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX
| |
Collapse
|
104
|
Sato K, Kennedy L, Liangpunsakul S, Kusumanchi P, Yang Z, Meng F, Glaser S, Francis H, Alpini G. Intercellular Communication between Hepatic Cells in Liver Diseases. Int J Mol Sci 2019; 20:2180. [PMID: 31052525 PMCID: PMC6540342 DOI: 10.3390/ijms20092180] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
Liver diseases are perpetuated by the orchestration of hepatocytes and other hepatic non-parenchymal cells. These cells communicate and regulate with each other by secreting mediators such as peptides, hormones, and cytokines. Extracellular vesicles (EVs), small particles secreted from cells, contain proteins, DNAs, and RNAs as cargos. EVs have attracted recent research interests since they can communicate information from donor cells to recipient cells thereby regulating physiological events via delivering of specific cargo mediators. Previous studies have demonstrated that liver cells secrete elevated numbers of EVs during diseased conditions, and those EVs are internalized into other liver cells inducing disease-related reactions such as inflammation, angiogenesis, and fibrogenesis. Reactions in recipient cells are caused by proteins and RNAs carried in disease-derived EVs. This review summarizes cell-to-cell communication especially via EVs in the pathogenesis of liver diseases and their potential as a novel therapeutic target.
Collapse
Grants
- TREATMENT OF PRIMARY SCLEROSING CHOLANGITIS USING EXTRACELLULAR VESICLES PSC Partners Seeking a Cure
- R01 DK110035 NIDDK NIH HHS
- I01 BX000574 BLRD VA
- 5I01BX000574, 1I01BX003031, 1I01BX001724, I01CX000361 VA Merit Award
- IK6 BX004601 BLRD VA
- R01 DK108959 NIDDK NIH HHS
- K01 AA026385 NIAAA NIH HHS
- I01 BX001724 BLRD VA
- DK054811, DK076898, DK107310, DK110035, DK062975, AA025997, DK108959, AA025208, DK107682, AA026917, AA026903, AA025157, and AA026385 NIH HHS
Collapse
Affiliation(s)
- Keisaku Sato
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lindsey Kennedy
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Suthat Liangpunsakul
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Praveen Kusumanchi
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Zhihong Yang
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Fanyin Meng
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University, Temple, TX 76504, USA.
| | - Heather Francis
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Gianfranco Alpini
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA.
- Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
105
|
Jiang K, Dong C, Yin Z, Li R, Wang Q, Wang L. The critical role of exosomes in tumor biology. J Cell Biochem 2019; 120:6820-6832. [PMID: 30362158 DOI: 10.1002/jcb.27813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/12/2018] [Indexed: 01/24/2023]
Abstract
Exosomes are a type of extracellular vesicles (diameter, 30-160 nm), which contain multiple proteins, nucleic acids, lipid molecules, and other substances. Most types of cells can secrete exosomes, although the biogenesis, composition, and function is specific to different cell types. Recently, many studies have demonstrated that exosomes play a critical role in tumor development. In this review, we briefly summarize the biogenesis, composition, and function of exosomes. We also discuss the recent advances in the critical role of exosomes in tumor biology with a special focus on their application in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Keqiu Jiang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, China
| | - Chengyong Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zeli Yin
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, China
| | - Rui Li
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, China
| | - Qi Wang
- Division of Respiratory Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liming Wang
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, China.,Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
106
|
Sato K, Glaser S, Kennedy L, Liangpunsakul S, Meng F, Francis H, Alpini G. Preclinical insights into cholangiopathies: disease modeling and emerging therapeutic targets. Expert Opin Ther Targets 2019; 23:461-472. [PMID: 30990740 DOI: 10.1080/14728222.2019.1608950] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The common predominant clinical features of cholangiopathies such as primary sclerosing cholangitis (PSC), primary biliary cholangitis (PBC), and biliary atresia (BA) are biliary damage/senescence and liver fibrosis. Curative therapies are lacking, and liver transplantation is the only option. An understanding of the mechanisms and pathogenesis is needed to develop novel therapies. Previous studies have developed various disease-based research models and have identified candidate therapeutic targets. Areas covered: This review summarizes recent studies performed in preclinical models of cholangiopathies and the current understanding of the pathophysiology representing potential targets for novel therapies. A literature search was conducted in PubMed using the combination of the searched term 'cholangiopathies' with one or two keywords including 'model', 'cholangiocyte', 'animal', or 'fibrosis'. Papers published within five years were obtained. Expert opinion: Access to appropriate research models is a key challenge in cholangiopathy research; establishing more appropriate models for PBC is an important goal. Several preclinical studies have demonstrated promising results and have led to novel therapeutic approaches, especially for PSC. Further studies on the pathophysiology of PBC and BA are necessary to identify candidate targets. Innovative therapeutic approaches such as stem cell transplantation have been introduced, and those therapies could be applied to PSC, PBC, and BA.
Collapse
Affiliation(s)
- Keisaku Sato
- a Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA.,b Richard L. Roudebush VA Medical Center , Indianapolis , IN , USA
| | - Shannon Glaser
- c Department of Medical Physiology , Texas A&M University Collage of Medicine , Temple , TX , USA
| | - Lindsey Kennedy
- a Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA.,b Richard L. Roudebush VA Medical Center , Indianapolis , IN , USA
| | - Suthat Liangpunsakul
- a Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA.,b Richard L. Roudebush VA Medical Center , Indianapolis , IN , USA
| | - Fanyin Meng
- a Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA.,b Richard L. Roudebush VA Medical Center , Indianapolis , IN , USA
| | - Heather Francis
- a Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA.,b Richard L. Roudebush VA Medical Center , Indianapolis , IN , USA
| | - Gianfranco Alpini
- a Indiana Center for Liver Research, Division of Gastroenterology & Hepatology, Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA.,b Richard L. Roudebush VA Medical Center , Indianapolis , IN , USA
| |
Collapse
|
107
|
Yin C, Han Q, Xu D, Zheng B, Zhao X, Zhang J. SALL4-mediated upregulation of exosomal miR-146a-5p drives T-cell exhaustion by M2 tumor-associated macrophages in HCC. Oncoimmunology 2019; 8:1601479. [PMID: 31143524 DOI: 10.1080/2162402x.2019.1601479] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/03/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence indicates that cancer cell-derived exosomes contribute to cancer progression through the modulation of tumor microenvironment, but the underlying mechanisms are not fully elucidated. Here, we reported that hepatocellular carcinoma (HCC)-derived exosomes could remodel macrophages by activating NF-κB signaling and inducing pro-inflammatory factors, and resulted in M2-polarized tumor-associated macrophages. In addition, the expression of IFN-γ and TNF-α was inhibited, while the expression of inhibitory receptors such as PD-1 and CTLA-4 was upregulated in T cells by HCC-derived exosome educated macrophages. Data also revealed that HCC exosomes were enriched with miR-146a-5p and promoted M2-polarization. Further investigation demonstrated that the transcription factor Sal-like protein-4 (SALL4) was critical for regulating miR-146a-5p in HCC exosomes and M2-polarization. Mechanistically, SALL4 could bind to the promoter of miR-146a-5p, and directly controlled its expression in exosomes. Blocking the SALL4/miR-146a-5p interaction in HCC reduced the expression of inhibitory receptors on T cells, reversed T cell exhaustion, and delayed HCC progression in DEN/CCL4-induced HCC mice. In conclusion, identification of a role of the exosomal SALL4/miR-146a-5p regulatory axis in M2-polarization as well as HCC progression provides potential targets for therapeutic and diagnostic applications in liver cancer.
Collapse
Affiliation(s)
- Chunlai Yin
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Dongqing Xu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Bingqing Zheng
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Xuemei Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
108
|
Ray RB, Ray R. Hepatitis C Virus Manipulates Humans as its Favorite Host for a Long-Term Relationship. Hepatology 2019; 69:889-900. [PMID: 30102776 PMCID: PMC6351149 DOI: 10.1002/hep.30214] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
Chronic hepatitis C virus (HCV) infection-associated liver disease is a global health problem. HCV often causes silent disease, and eventually progresses to end-stage liver disease. HCV infects hepatocytes; however, initial manifestation of liver disease is mostly displayed in hepatic stellate cells (HSCs), causing fibrosis/cirrhosis, and is believed to occur from inflammation in the liver. It remains unclear why HCV is not spontaneously cleared from infected liver in the majority of individuals and develops chronic infection with progressive liver disease. Direct-acting antivirals (DAAs) show excellent results in controlling viremia, although beneficial consequence in advanced liver disease remains to be understood. In this review, we highlight the current knowledge that has contributed to our understanding of the role of HCV in inflammation, immune evasion, metabolic disorders, liver pathogeneses, and efforts in vaccine development.
Collapse
Affiliation(s)
- Ratna B. Ray
- Department of Pathology, Saint Louis University, Saint Louis, Missouri 63104, USA,Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri 63104, USA
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri 63104, USA
| |
Collapse
|
109
|
The utility of serum exosomal microRNAs in hepatocellular carcinoma. Biomed Pharmacother 2019; 111:1221-1227. [PMID: 30841435 DOI: 10.1016/j.biopha.2018.12.131] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/26/2018] [Accepted: 12/30/2018] [Indexed: 12/31/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of cancer worldwide and the third most common cause of cancer-associated deaths each year. Due to the invasive, fast growth and insidious onset of HCC, most patients are diagnosed at an advanced stage, reducing the effectiveness of radical surgery. Therefore, effective early detection and diagnostic methods are crucial for improving the treatment and prognosis of HCC patients. Exosomes are 40- to 100-nm-sized vesicles that are released from many cell types into the extracellular space, and extensive evidence has suggested that exosomes are involved in the occurrence and development of HCC. Recently, the presence of microRNAs (miRNAs) in exosomes has been verified. Such miRNAs can be internalized by neighboring or distant cells to subsequently regulate multiple target genes in recipient cells at the posttranscriptional level and affect the processes of cell proliferation, differentiation and apoptosis. Herein, we summarize the current knowledge about the potential utility of serum exosomal miRNAs in the diagnosis and treatment of HCC.
Collapse
|
110
|
Yao Z, Jia X, Megger DA, Chen J, Liu Y, Li J, Sitek B, Yuan Z. Label-Free Proteomic Analysis of Exosomes Secreted from THP-1-Derived Macrophages Treated with IFN-α Identifies Antiviral Proteins Enriched in Exosomes. J Proteome Res 2018; 18:855-864. [DOI: 10.1021/acs.jproteome.8b00514] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zhenlan Yao
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Xiaofang Jia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Dominik A. Megger
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 50 Virchowstraße 179, 45147 Essen, Germany
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yuyi Liu
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jianhua Li
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| |
Collapse
|
111
|
Liu CH, Ampuero J, Gil-Gómez A, Montero-Vallejo R, Rojas Á, Muñoz-Hernández R, Gallego-Durán R, Romero-Gómez M. miRNAs in patients with non-alcoholic fatty liver disease: A systematic review and meta-analysis. J Hepatol 2018; 69:1335-1348. [PMID: 30142428 DOI: 10.1016/j.jhep.2018.08.008] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/21/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS microRNAs (miRNAs) are deregulated in non-alcoholic fatty liver disease (NAFLD) and have been proposed as useful markers for the diagnosis and stratification of disease severity. We conducted a meta-analysis to identify the potential usefulness of miRNA biomarkers in the diagnosis and stratification of NAFLD severity. METHODS After a systematic review, circulating miRNA expression consistency and mean fold-changes were analysed using a vote-counting strategy. The sensitivity, specificity, positive and negative likelihood ratios, diagnostic odds ratio and area under the curve (AUC) for the diagnosis of NAFLD or non-alcoholic steatohepatitis (NASH) were pooled using a bivariate meta-analysis. Deeks' funnel plot was used to assess the publication bias. RESULTS Thirty-seven studies of miRNA expression profiles and six studies of diagnostic accuracy were ultimately included in the quantitative analysis. miRNA-122 and miRNA-192 showed consistent upregulation. miRNA-122 was upregulated in every scenario used to distinguish NAFLD severity. The miRNA expression correlation between the serum and liver tissue was inconsistent across studies. miRNA-122 distinguished NAFLD from healthy controls with an AUC of 0.82 (95% CI 0.75-0.89), and miRNA-34a distinguished non-alcoholic steatohepatitis (NASH) from non-alcoholic fatty liver (NAFL) with an AUC of 0.78 (95% CI 0.67-0.88). CONCLUSION miRNA-34a, miRNA-122 and miRNA-192 were identified as potential diagnostic markers to segregate NAFL from NASH. Both miRNA-122, in distinguishing NAFLD from healthy controls, and miRNA-34a, in distinguishing NASH from NAFL, showed moderate diagnostic accuracy. miRNA-122 was upregulated in every scenario of NAFL, NASH and fibrosis. LAY SUMMARY: microRNAs are deregulated in non-alcoholic fatty liver disease. The microRNAs, miRNA-34a, miRNA-122 and miRNA-192, were identified as potential biomarkers of non-alcoholic fatty liver and non-alcoholic steatohepatitis, at different stages of disease severity. The correlation between miRNA expression in the serum and in liver tissue was inconsistent, or even inverse.
Collapse
Affiliation(s)
- Chang-Hai Liu
- Institute of Biomedicine of Seville, Sevilla, Spain; University of Seville, Seville, Spain
| | - Javier Ampuero
- Institute of Biomedicine of Seville, Sevilla, Spain; Unit of Digestive Diseases and Ciberehd, University Hospital Virgen del Rocío, Seville, Spain; University of Seville, Seville, Spain
| | - Antonio Gil-Gómez
- Institute of Biomedicine of Seville, Sevilla, Spain; University of Seville, Seville, Spain
| | - Rocío Montero-Vallejo
- Institute of Biomedicine of Seville, Sevilla, Spain; University of Seville, Seville, Spain
| | - Ángela Rojas
- Institute of Biomedicine of Seville, Sevilla, Spain
| | | | | | - Manuel Romero-Gómez
- Institute of Biomedicine of Seville, Sevilla, Spain; Unit of Digestive Diseases and Ciberehd, University Hospital Virgen del Rocío, Seville, Spain; University of Seville, Seville, Spain.
| |
Collapse
|
112
|
Sung S, Kim J, Jung Y. Liver-Derived Exosomes and Their Implications in Liver Pathobiology. Int J Mol Sci 2018; 19:3715. [PMID: 30469540 PMCID: PMC6320937 DOI: 10.3390/ijms19123715] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 02/08/2023] Open
Abstract
The liver has a wide range of physiological functions in the body, and its health is maintained by complex cross-talk among hepatic cells, including parenchymal hepatocytes and nonparenchymal cells. Exosomes, which are one method of cellular communication, are endosomal-derived small vesicles that are released by donor cells and delivered to the target cells at both short and long distances. Because exosomes carry a variety of cargoes, including proteins, mRNAs, microRNAs and other noncoding RNAs originating from donor cells, exosomes convey cellular information that enables them to potentially serve as biomarkers and therapeutics in liver diseases. Hepatocytes release exosomes to neighboring hepatocytes or nonparenchymal cells to regulate liver regeneration and repair. Nonparenchymal cells, including hepatic stellate cells, liver sinusoidal endothelial cells, and cholangiocytes, also secrete exosomes to regulate liver remodeling upon liver injury. Exosomes that are released from liver cancer cells create a favorable microenvironment for cancer growth and progression. In this review, we summarize and discuss the current findings and understanding of exosome-mediated intercellular communication in the liver, with a particular focus on the function of exosomes in both health and disease. Based on the current findings, we suggest the potential applications of exosomes as biomarkers and therapeutics for liver diseases.
Collapse
Affiliation(s)
- Sumi Sung
- Department of Integrated Biological Science, Pusan National University, 63-2 Pusandaehak-ro, Kumjeong-gu, Pusan 46241, Korea.
| | - Jieun Kim
- Department of Integrated Biological Science, Pusan National University, 63-2 Pusandaehak-ro, Kumjeong-gu, Pusan 46241, Korea.
| | - Youngmi Jung
- Department of Integrated Biological Science, Pusan National University, 63-2 Pusandaehak-ro, Kumjeong-gu, Pusan 46241, Korea.
- Department of Biological Sciences, Pusan National University, 63-2 Pusandaehak-ro, Kumjeong-gu, Pusan 46241, Korea.
| |
Collapse
|
113
|
Abdelrahma F, Alkarim S, Mohamed Sa I. Ameliorative Effect of Mesenchymal Stem Cells-derived Exosomes on Diethylnitrosamine-induced Liver Injury in Albino Rats. INT J PHARMACOL 2018. [DOI: 10.3923/ijp.2018.1128.1135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
114
|
Morán L, Cubero FJ. Extracellular vesicles in liver disease and beyond. World J Gastroenterol 2018; 24:4519-4526. [PMID: 30386101 PMCID: PMC6209575 DOI: 10.3748/wjg.v24.i40.4519] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/02/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane-derived vesicles which can be released by different cell types, including hepatocytes, hepatic stellate cells and immune cells in normal and pathological conditions. EVs carry lipids, proteins, coding and non-coding RNAs and mitochondrial DNA causing modifications on the recipient cells. These vesicles are considered potential biomarkers and therapeutic agents for human diagnostic and prognostic due to their function as intercellular mediators of cell-cell communication within the liver and between other organs. However, the development and optimization of methods for EVs isolation is required to characterize their biological functions as well as their potential as a treatment option in the clinic. Nevertheless, many questions remain unanswered related to the function of EVs under physiological and pathological conditions. In the current editorial, the results obtained in different studies that investigated the role of intrahepatic EVs during liver diseases, including drug-induced liver injury, non-alcoholic fatty liver, non-alcoholic steatohepatitis, alcoholic liver disease and hepatocellular carcinoma and extrahepatic EVs in remote organs during pathological events such as pulmonary disease, cardiovascular diseases, neurodegenerative disorders e.g., Alzheimer’s disease, Parkinson’s disease and multiple sclerosis as well as in immunopathological processes, are discussed. Although much light needs to be shed on the mechanisms of EVs, these membrane-derived vesicles represent both a novel promising diagnostic, and a therapeutic tool for clinical use that we emphasize in the current editorial.
Collapse
Affiliation(s)
- Laura Morán
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| |
Collapse
|
115
|
Fang JH, Zhang ZJ, Shang LR, Luo YW, Lin YF, Yuan Y, Zhuang SM. Hepatoma cell-secreted exosomal microRNA-103 increases vascular permeability and promotes metastasis by targeting junction proteins. Hepatology 2018; 68:1459-1475. [PMID: 29637568 DOI: 10.1002/hep.29920] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 03/20/2018] [Accepted: 03/30/2018] [Indexed: 12/11/2022]
Abstract
UNLABELLED Increased vascular permeability facilitates metastasis. Emerging evidence indicates that secreted microRNAs (miRNAs) may mediate the crosstalk between cancer and stromal cells. To date, whether and how secreted miRNAs affect vascular permeability remains unclear. Based on deep sequencing and quantitative PCR, we found that higher level of serum miR-103 was associated with higher metastasis potential of hepatocellular carcinoma (HCC). The in vitro endothelial permeability and transendothelial invasion assays revealed that the conditioned media or exosomes derived from high miR-103-expressing hepatoma cells increased the permeability of endothelial monolayers, but this effect was attenuated if exosome secretion of hepatoma cells was blocked by silencing ALIX and HRS or if miR-103 within hepatoma or endothelial cells was antagonized. Most importantly, pretreating endothelial monolayers with exosomes that were from stable miR-103-expressing hepatoma cells facilitated the transendothelial invasion of tumor cells, and this role of exosomes was abrogated by inhibiting miR-103 in endothelial cells. Further in vivo analyses disclosed that mice with xenografts of stable miR-103-expressing hepatoma cells exhibited higher vascular permeability in tumor, higher level of exosomal miR-103 and greater number of tumor cells in blood circulation, and increased rates of hepatic and pulmonary metastases, compared to control mice. Mechanism investigations revealed that hepatoma cell-secreted miR-103 could be delivered into endothelial cells via exosomes, and then attenuated the endothelial junction integrity by directly inhibiting the expression of VE-Cadherin (VE-Cad), p120-catenin (p120) and zonula occludens 1. Moreover, miR-103 could also promote tumor cell migration by repressing p120 expression in hepatoma cells. CONCLUSION Hepatoma cell-secreted exosomal miR-103 increases vascular permeability and promotes tumor metastasis by targeting multiple endothelial junction proteins, which highlights secreted miR-103 as a potential therapeutic target and a predictive marker for HCC metastasis. (Hepatology 2018).
Collapse
Affiliation(s)
- Jian-Hong Fang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zi-Jun Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Li-Ru Shang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yu-Wei Luo
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yi-Fang Lin
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yunfei Yuan
- Department of Hepatobilliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Shi-Mei Zhuang
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
116
|
Zhang C, Yang X, Qi Q, Gao Y, Wei Q, Han S. lncRNA-HEIH in serum and exosomes as a potential biomarker in the HCV-related hepatocellular carcinoma. Cancer Biomark 2018; 21:651-659. [PMID: 29286922 DOI: 10.3233/cbm-170727] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chronic hepatitis C (CHC) is a contagious liver disease that results from infection with the hepatitis C virus (HCV). The most serious consequence of CHC is HCV-related hepatocellular carcinoma (HCC). OBJECTIVE To illustrate the clinical significance of lncRNA HEIH expression in serum and exosomes in the development of HCV-related HCC. METHODS Thirty-five CHC, twenty-two HCV-induced cirrhosis and ten HCV-related HCC patients in Huzhou Central Hospital from January 2016 to September 2016 were recruited in the present study. Basic patient information, clinical serological indicators, and clinical imaging data were investigated and analyzed. Serum samples were collected from patients after receiving informed consent. Exosomes were extracted from the serum, and electron microscopy was used to observe the ultrastructure of exosomes. Quantitative PCR was used to detect lncRNA HEIH gene expression in serum and exosomes. RESULTS The changes in the ALT, GGT, HDL, INR, Alb and AFP levels in the patients with HCV-induced cirrhosis and HCV-related HCC were statistically significant. In patients with HCV-related HCC, lncRNA-HEIH expression in serum and exosomes was increased, but the ratio of lncRNA-HEIH expression in serum versus exosomes was decreased compared to patients with CHC.
Collapse
Affiliation(s)
- Chun Zhang
- Department of Infectious Diseases, First Hospital Affiliated to Suzhou University, Suzhou, Jiangsu 215006, China
| | - Xi Yang
- Department of Intervention and Radiotherapy, Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Quan Qi
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Yuhai Gao
- Department of General Surgery, Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Qiang Wei
- Department of General Surgery, Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| | - Shuwen Han
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang 313000, China
| |
Collapse
|
117
|
Schulze S, Stöß C, Lu M, Wang B, Laschinger M, Steiger K, Altmayr F, Friess H, Hartmann D, Holzmann B, Hüser N. Cytosolic nucleic acid sensors of the innate immune system promote liver regeneration after partial hepatectomy. Sci Rep 2018; 8:12271. [PMID: 30115978 PMCID: PMC6095902 DOI: 10.1038/s41598-018-29924-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 07/20/2018] [Indexed: 12/19/2022] Open
Abstract
Stimulation of cytosolic nucleic acid sensors of innate immunity by pathogen-derived nucleic acids is important for antimicrobial defence, but stimulation through self-derived nucleic acids may contribute to autoinflammation and cancer. DNA sensing in the cytosol requires the stimulator of interferon genes (STING), while cytosolic RNA sensors use mitochondrial antiviral-signalling protein (MAVS). In a murine model of two-thirds hepatectomy, combined deficiency of MAVS and STING resulted in strongly impaired hepatocyte proliferation and delayed recovery of liver mass. Whereas lack of MAVS and STING did not influence upregulation of the G1-phase cyclins D1 and E1, it substantially reduced the hyperphosphorylation of retinoblastoma protein, attenuated the activation of cyclin-dependent kinase (CDK)-2, delayed upregulation of CDK1 and cyclins A2 and B1, and impaired S-phase entry of hepatocytes. Mechanistically, lack of cytosolic nucleic acid sensors strongly upregulated the anti-proliferative mediators TGF-β2 and activin A, which was associated with an increased expression of the cell cycle inhibitors p15 and p21. Partial hepatectomy was followed by the release of exosomes with abundant nucleic acid cargo, which may provide ligands for the MAVS and STING pathways. Together, these findings identify a previously unrecognised function of cytosolic nucleic acid sensors of innate immunity for promoting liver regeneration.
Collapse
Affiliation(s)
- Sarah Schulze
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Christian Stöß
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Miao Lu
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Baocai Wang
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Melanie Laschinger
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Katja Steiger
- Technical University of Munich, School of Medicine, Comparative Experimental Pathology, Institute of Pathology, Trogerstr. 18, 81675, Munich, Germany
| | - Felicitas Altmayr
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Helmut Friess
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Daniel Hartmann
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| | - Bernhard Holzmann
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Norbert Hüser
- Technical University of Munich, School of Medicine, Department of Surgery, Ismaninger Str. 22, 81675, Munich, Germany
| |
Collapse
|
118
|
Liang J, Zhang X, He S, Miao Y, Wu N, Li J, Gan Y. Sphk2 RNAi nanoparticles suppress tumor growth via downregulating cancer cell derived exosomal microRNA. J Control Release 2018; 286:348-357. [PMID: 30077738 DOI: 10.1016/j.jconrel.2018.07.039] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/21/2022]
Abstract
Exosomes secreted from cancer cells promote tumor progression through the transfection of containing microRNA (miRNA), mRNAs and proteins. Yet, little of this knowledge has translated into the therapeutic application. Herein, we propose a tumor therapeutic strategy via decreasing exosomal miRNA secretion. The study designed small interfering RNA (siRNA) loaded nanoparticles to downregulate sphingosine kinase 2 (Sphk2) and investigate their potential in decreasing exosomal oncogenic miRNA content and inhibiting tumor growth. The synthesized lipid (2E)-4-(dioleostearin)-amino-4‑carbonyl-2-butenoic (DC) and chitosan were utilized to produce siRNA loaded nanoparticles (DC/CS-siRNA NPs), with optimal siRNA complexation and high transfection efficacy. We demonstrated that Sphk2 gene silencing induced by nanoparticles in hepatocellular carcinoma (HCC) cells could reduce miRNA-21 sorting into exosomes, contributing to the inhibition of tumor cell migration and tumorigenic function of exosomes to normal liver cells. Furthermore, in xenograft mouse model, Sphk2 siRNA loaded DC/CS NPs could significantly block tumor progression of malignancy HCC. These results suggest a new therapeutic approach for tumor treatment by ablating oncogenic miRNA in malicious exosomes.
Collapse
Affiliation(s)
- Jinying Liang
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China; Development of pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Xinxin Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | - Shufang He
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | - Yunqiu Miao
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China
| | - Na Wu
- Development of pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Juan Li
- Development of pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai 201203, China.
| |
Collapse
|
119
|
Kostallari E, Hirsova P, Prasnicka A, Verma VK, Yaqoob U, Wongjarupong N, Roberts LR, Shah VH. Hepatic stellate cell-derived platelet-derived growth factor receptor-alpha-enriched extracellular vesicles promote liver fibrosis in mice through SHP2. Hepatology 2018; 68:333-348. [PMID: 29360139 PMCID: PMC6033667 DOI: 10.1002/hep.29803] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/11/2022]
Abstract
UNLABELLED Liver fibrosis is characterized by the activation and migration of hepatic stellate cells (HSCs), followed by matrix deposition. Recently, several studies have shown the importance of extracellular vesicles (EVs) derived from liver cells, such as hepatocytes and endothelial cells, in liver pathobiology. While most of the studies describe how liver cells modulate HSC behavior, an important gap exists in the understanding of HSC-derived signals and more specifically HSC-derived EVs in liver fibrosis. Here, we investigated the molecules released through HSC-derived EVs, the mechanism of their release, and the role of these EVs in fibrosis. Mass spectrometric analysis showed that platelet-derived growth factor (PDGF) receptor-alpha (PDGFRα) was enriched in EVs derived from PDGF-BB-treated HSCs. Moreover, patients with liver fibrosis had increased PDGFRα levels in serum EVs compared to healthy individuals. Mechanistically, in vitro tyrosine720-to-phenylalanine mutation on the PDGFRα sequence abolished enrichment of PDGFRα in EVs and redirected the receptor toward degradation. Congruently, the inhibition of Src homology 2 domain tyrosine phosphatase 2, the regulatory binding partner of phosphorylated tyrosine720, also inhibited PDGFRα enrichment in EVs. EVs derived from PDGFRα-overexpressing cells promoted in vitro HSC migration and in vivo liver fibrosis. Finally, administration of Src homology 2 domain tyrosine phosphatase 2inhibitor, SHP099, to carbon tetrachloride-administered mice inhibited PDGFRα enrichment in serum EVs and reduced liver fibrosis. CONCLUSION PDGFRα is enriched in EVs derived from PDGF-BB-treated HSCs in an Src homology 2 domain tyrosine phosphatase 2-dependent manner and these PDGFRα-enriched EVs participate in development of liver fibrosis. (Hepatology 2018;68:333-348).
Collapse
Affiliation(s)
- Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Alena Prasnicka
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN,Department of Pharmacology, Charles University, Hradec Kralove, Czech Republic
| | - Vikas K. Verma
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Usman Yaqoob
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
120
|
Liu J, Jiang M, Deng S, Lu J, Huang H, Zhang Y, Gong P, Shen X, Ruan H, Jin M, Wang H. miR-93-5p-Containing Exosomes Treatment Attenuates Acute Myocardial Infarction-Induced Myocardial Damage. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:103-115. [PMID: 29858047 PMCID: PMC5852413 DOI: 10.1016/j.omtn.2018.01.010] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 02/07/2023]
Abstract
Adipose-derived stromal cells (ADSCs) have been considered as an attractive therapeutic tool. Accumulating evidence indicates that the healing effects of ADSCs are mainly related to paracrine action rather than transdifferentiation. Data show that the expression of miR-93-5p has a cardio-protective effect after acute myocardial infarction (AMI). To identify whether miR-93-5p-encapsulating exosomes that form ADSCs have a better cardio-protective effect, we investigated the inflammatory factors and miR-30d-5p expression in clinical levels. A rat model of AMI and an in vitro model of hypoxic H9c2 cells were established to study the protective mechanism of miR-93-5p in ischemia-induced cardiac injury. The results show that the expression of inflammatory cytokines and miR-93-5p were increased following AMI in both patients and animal models. Moreover, treatment with ADSC-derived miR-93-5p-containing exosomes has a greater protective effect on infarction-induced myocardial damage than simple exosome processing. Furthermore, in vitro experiments confirmed that the expression of miR-93-5p can significantly suppress hypoxia-induced autophagy and inflammatory cytokine expression by targeting Atg7 and Toll-like receptor 4 (TLR4), respectively, and was confirmed with Atg7 or TLR4 overexpression. The results also show that autophagy activation can promote inflammatory cytokine expression indirectly. Taken together, these results suggest that the miR-93-5p-enhanced ADSC-derived exosomes prevent cardiac injury by inhibiting autophagy and the inflammatory response.
Collapse
Affiliation(s)
- Jiwen Liu
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Mei Jiang
- Department of Neurology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Shengqiong Deng
- Department of Clinical Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Jide Lu
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Hui Huang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Yu Zhang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Peihua Gong
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Xumin Shen
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Huanjun Ruan
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China
| | - Mingming Jin
- Department of Central Laboratory, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China.
| | - Hairong Wang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai 200135, China.
| |
Collapse
|
121
|
Qiu Y, Wang S, Wan T, Ye M, Jiang R, Pei L, Yang L. Blood-based novel biomarkers for nonalcoholic steatohepatitis. Biomark Med 2018; 12:501-515. [PMID: 29712439 DOI: 10.2217/bmm-2017-0361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Nonalcoholic fatty liver disease has become a social health challenge of global concern. The term nonalcoholic steatohepatitis (NASH) is a more severe condition than simple steatosis and distinguishing NASH from nonalcoholic fatty liver disease is particularly important. Liver biopsy remains a gold standard in diagnosing NASH. Meanwhile, radiological techniques such as ultrasonography and MRI are also applied widely. However, the invasive and expensive examination is not suitable for screening, and there is a great need for reliable and appropriate biomarkers to screen patients for NASH. Based on the current studies of blood-based novel biomarkers, we attempt to summarize the latest findings on biomarkers for NASH, including blood biomarkers encompassing proteins, lipids and miRNAs; the correlation between extracellular vesicles and NASH; and treatment strategies for NASH.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Sufan Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Ting Wan
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Mingtong Ye
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Rui Jiang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Lei Pei
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| | - Lili Yang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.,Guangdong Provincial Key Laboratory of Food, Nutrition & Health, Guangzhou, Guangdong 510080, PR China
| |
Collapse
|
122
|
Olaizola P, Lee-Law PY, Arbelaiz A, Lapitz A, Perugorria MJ, Bujanda L, Banales JM. MicroRNAs and extracellular vesicles in cholangiopathies. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1293-1307. [PMID: 28711597 DOI: 10.1016/j.bbadis.2017.06.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 12/22/2022]
Abstract
UNLABELLED Cholangiopathies encompass a heterogeneous group of disorders affecting biliary epithelial cells (i.e. cholangiocytes). Early diagnosis, prognosis and treatment still remain clinically challenging for most of these diseases and are critical for adequate patient care. In the past decade, extensive research has emphasized microRNAs (miRs) as potential non-invasive biomarkers and tools to accurately identify, predict and treat cholangiopathies. MiRs can be released extracellularly conjugated with lipoproteins or encapsulated in extracellular vesicles (EVs). Research on EVs is also gaining attention since they are present in multiple biological fluids and may represent a relevant source of novel non-invasive biomarkers and be vehicles for new therapeutic approaches. This review highlights the most promising candidate miRs and EV-related biomarkers in cholangiopathies, as well as their relevant roles in biliary pathophysiology. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen. RESEARCH STRATEGY PubMed search (April 2017) was done with the following terms: "microRNA", "miRNA", "miR", "extracellular vesicles", "EV", "exosomes", "primary biliary cholangitis", "primary biliary cholangitis", "PBC", "primary sclerosing cholangitis", "PSC", "cholangiocarcinoma", "CCA", "biliary atresia", "BA", "polycystic liver diseases", "PLD", "cholangiopathies", "cholestatic liver disease". Most significant articles in full-text English were selected. The reference lists of selected papers were also considered.
Collapse
Affiliation(s)
- P Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - P Y Lee-Law
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; Department of Gastroenterology and Hepatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - A Arbelaiz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - A Lapitz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - M J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - L Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - J M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
123
|
Sato K, Meng F, Giang T, Glaser S, Alpini G. Mechanisms of cholangiocyte responses to injury. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1262-1269. [PMID: 28648950 PMCID: PMC5742086 DOI: 10.1016/j.bbadis.2017.06.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/06/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022]
Abstract
Cholangiocytes, epithelial cells that line the biliary epithelium, are the primary target cells for cholangiopathies including primary sclerosing cholangitis and primary biliary cholangitis. Quiescent cholangiocytes respond to biliary damage and acquire an activated neuroendocrine phenotype to maintain the homeostasis of the liver. The typical response of cholangiocytes is proliferation leading to bile duct hyperplasia, which is a characteristic of cholestatic liver diseases. Current studies have identified various signaling pathways that are associated with cholangiocyte proliferation/loss and liver fibrosis in cholangiopathies using human samples and rodent models. Although recent studies have demonstrated that extracellular vesicles and microRNAs could be mediators that regulate these messenger/receptor axes, further studies are required to confirm their roles. This review summarizes current studies of biliary response and cholangiocyte proliferation during cholestatic liver injury with particular emphasis on the secretin/secretin receptor axis. This article is part of a Special Issue entitled: Cholangiocytes in Health and Diseaseedited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Collapse
Affiliation(s)
- Keisaku Sato
- Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, TX, United States; Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, United States; Academic Research Integration, Baylor Scott & White Health, Temple, TX, United States; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | - Thao Giang
- Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, TX, United States; Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, United States; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, United States
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, TX, United States; Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Temple, TX, United States; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, TX, United States.
| |
Collapse
|
124
|
Exosome-Mimetic Nanovesicles from Hepatocytes promote hepatocyte proliferation in vitro and liver regeneration in vivo. Sci Rep 2018; 8:2471. [PMID: 29410409 PMCID: PMC5802835 DOI: 10.1038/s41598-018-20505-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
The liver has great regenerative capacity after functional mass loss caused by injury or disease. Many studies have shown that primary hepatocyte-derived exosomes, which can deliver biological information between cells, promote the regenerative process of the liver. However, the yield of exosomes is very limited. Recent studies have demonstrated that exosome-mimetic nanovesicles (NVs) can be prepared from cells with almost 100 times the production yield compared with exosomes. Thus, this study investigated the therapeutic capacity of exosome-mimetic NVs from primary hepatocytes in liver regeneration. Exosome-mimetic NVs were prepared by serial extrusions of cells through polycarbonate membranes, and the yield of these NVs was more than 100 times that of exosomes. The data indicated that the NVs could promote hepatocyte proliferation and liver regeneration by significantly enhancing the content of sphingosine kinase 2 in recipient cells. To the best of our knowledge, this is the first time that exosome-mimetic NVs from primary hepatocytes have been prepared, and these NVs have components similar to exosomes from primary hepatocytes and, in some respects, biofunctions similar to exosomes. Strategies inspired by this study may lead to substitution of exosomes with exosome-mimetic NVs for biofunctional purposes, including utilization in tissue repair and regeneration.
Collapse
|
125
|
Obesity linking to hepatocellular carcinoma: A global view. Biochim Biophys Acta Rev Cancer 2018; 1869:97-102. [PMID: 29366974 DOI: 10.1016/j.bbcan.2017.12.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/20/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is the commonest primary liver cancer and the second leading cause of cancer death worldwide. Obesity is rapidly becoming pandemic and associated with increased carcinogenesis. In this review, we describe the obesity-related factors that influence the development of HCC. We provide evidence of strong links between neural regulation, endocrine and HCC in obesity. We discuss recent advances in our understanding of how adipose tissue alters hepatic metabolism and immune response in HCC development through inter-organ communication. Taken together, our review aims to provides a concise and up-to date summary about the connection between obesity and HCC, with emphasis on the opportunities for effective strategies in preventing the development of HCC in obese individuals.
Collapse
|
126
|
Lee CW, Chen YF, Wu HH, Lee OK. Historical Perspectives and Advances in Mesenchymal Stem Cell Research for the Treatment of Liver Diseases. Gastroenterology 2018; 154:46-56. [PMID: 29107021 DOI: 10.1053/j.gastro.2017.09.049] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 09/23/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022]
Abstract
Liver transplantation is the only effective therapy for patients with decompensated cirrhosis and fulminant liver failure. However, due to a shortage of donor livers and complications associated with immune suppression, there is an urgent need for new therapeutic strategies for patients with end-stage liver diseases. Given their unique function in self-renewal and differentiation potential, stem cells might be used to regenerate damaged liver tissue. Recent studies have shown that stem cell-based therapies can improve liver function in a mouse model of hepatic failure. Moreover, acellular liver scaffolds seeded with hepatocytes produced functional bioengineered livers for organ transplantation in preclinical studies. The therapeutic potential of stem cells or their differentiated progenies will depend on their capacity to differentiate into mature and functional cell types after transplantation. It will also be important to devise methods to overcome their genomic instability, immune reactivity, and tumorigenic potential. We review directions and advances in the use of mesenchymal stem cells and their derived hepatocytes for liver regeneration. We also discuss the potential applications of hepatocytes derived from human pluripotent stem cells and challenges to using these cells in treating end-stage liver disease.
Collapse
Affiliation(s)
- Chien-Wei Lee
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Fan Chen
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Hsiang Wu
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Oscar K Lee
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Taipei City Hospital, Taipei, Taiwan.
| |
Collapse
|
127
|
Samoylova EM, Kalsin VA, Bespalova VA, Devichensky VM, Baklaushev VP. Exosomes: from biology to clinics. GENES & CELLS 2017; 12:7-19. [DOI: 10.23868/201707024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2025]
Abstract
Exosomes are extracellular vesicles with the diameter of 30-120 nm, originating from early endosomes. Exosomes have been actively studied in the last decade, and a great amount of data has appeared on their nature and role in the intercellular transport and signaling both in the normal and pathological conditions. A particular interest to exosomes in the clinical practice emerged after the separation of their circulating fraction from the blood and the study of tumor genetic markers in them became possible (so called “liquid biopsy”). The objective of this review is to familiarize clinical specialists with the fundamentals of exosomes' biology and physiology and with the main achievements on their practical application in the medicine, as a natural drug delivery system, as well as for high-precision, early non-invasive differential diagnostics of diseases.
Collapse
|
128
|
Alvaro D. The challenge of cholangiocarcinoma diagnosis: The turning point is in extracellular vesicles? Hepatology 2017; 66:1029-1031. [PMID: 28605030 DOI: 10.1002/hep.29314] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 06/04/2017] [Accepted: 06/06/2017] [Indexed: 12/29/2022]
Affiliation(s)
- Domenico Alvaro
- Division of Gastroenterology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
129
|
Kim SH, Kim G, Han DH, Lee M, Kim I, Kim B, Kim KH, Song YM, Yoo JE, Wang HJ, Bae SH, Lee YH, Lee BW, Kang ES, Cha BS, Lee MS. Ezetimibe ameliorates steatohepatitis via AMP activated protein kinase-TFEB-mediated activation of autophagy and NLRP3 inflammasome inhibition. Autophagy 2017; 13:1767-1781. [PMID: 28933629 DOI: 10.1080/15548627.2017.1356977] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Impairment in macroautophagy/autophagy flux and inflammasome activation are common characteristics of nonalcoholic steatohepatitis (NASH). Considering the lack of approved agents for treating NASH, drugs that can enhance autophagy and modulate inflammasome pathways may be beneficial. Here, we investigated the novel mechanism of ezetimibe, a widely prescribed drug for hypercholesterolemia, as a therapeutic option for ameliorating NASH. Human liver samples with steatosis and NASH were analyzed. For in vitro studies of autophagy and inflammasomes, primary mouse hepatocytes, human hepatoma cells, mouse embryonic fibroblasts with Ampk or Tsc2 knockout, and human or primary mouse macrophages were treated with ezetimibe and palmitate. Steatohepatitis and fibrosis were induced by feeding Atg7 wild-type, haploinsufficient, and knockout mice a methionine- and choline-deficient diet with ezetimibe (10 mg/kg) for 4 wk. Human livers with steatosis or NASH presented impaired autophagy with decreased nuclear TFEB and increased SQSTM1, MAP1LC3-II, and NLRP3 expression. Ezetimibe increased autophagy flux and concomitantly ameliorated lipid accumulation and apoptosis in palmitate-exposed hepatocytes. Ezetimibe induced AMPK phosphorylation and subsequent TFEB nuclear translocation, related to MAPK/ERK. In macrophages, ezetimibe blocked the NLRP3 inflammasome-IL1B pathway in an autophagy-dependent manner and modulated hepatocyte-macrophage interaction via extracellular vesicles. Ezetimibe attenuated lipid accumulation, inflammation, and fibrosis in liver-specific Atg7 wild-type and haploinsufficient mice, but not in knockout mice. Ezetimibe ameliorates steatohepatitis by autophagy induction through AMPK activation and TFEB nuclear translocation, related to an independent MTOR ameliorative effect and the MAPK/ERK pathway. Ezetimibe dampens NLRP3 inflammasome activation in macrophages by modulating autophagy and a hepatocyte-driven exosome pathway.
Collapse
Affiliation(s)
- Soo Hyun Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Gyuri Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,b Department of Medicine, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea
| | - Dai Hoon Han
- d Department of Surgery , Yonsei University College of Medicine , Seoul , Korea
| | - Milim Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Irene Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Bohkyung Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Kook Hwan Kim
- e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Young-Mi Song
- f Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital , University of Toronto , Toronto , Canada
| | - Jeong Eun Yoo
- g Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| | - Hye Jin Wang
- h Department of Pharmacology , Yonsei University College of Medicine , Seoul , Korea
| | - Soo Han Bae
- e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Yong-Ho Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Byung-Wan Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Eun Seok Kang
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Bong-Soo Cha
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Myung-Shik Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| |
Collapse
|
130
|
Sato K, Meng F, Venter J, Giang T, Glaser S, Alpini G. The role of the secretin/secretin receptor axis in inflammatory cholangiocyte communication via extracellular vesicles. Sci Rep 2017; 7:11183. [PMID: 28894209 PMCID: PMC5593902 DOI: 10.1038/s41598-017-10694-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022] Open
Abstract
Small and large intrahepatic bile ducts consist of small and large cholangiocytes, respectively, and these cholangiocytes have different morphology and functions. The gastrointestinal peptide hormone, secretin (SCT) that binds to secretin receptor (SR), is a key mediator in cholangiocyte pathophysiology. Extracellular vesicles (EVs) are membrane-bound vesicles and cell-cell EV communication is recognized as an important factor in liver pathology, although EV communication between cholangiocytes is not identified to date. Cholangiocytes secrete proinflammatory cytokines during bacterial infection leading to biliary inflammation and hyperplasia. We demonstrate that cholangiocytes stimulated with lipopolysaccharide (LPS), which is a membrane component of gram-negative bacteria, secrete more EVs than cholangiocytes incubated with vehicle. These LPS-derived EVs induce inflammatory responses in other cholangiocytes including elevated cytokine production and cell proliferation. Large but not small cholangiocytes show inflammatory responses against large but not small cholangiocyte-derived EVs. Large cholangiocytes with knocked down either SCT or SR by short hairpin RNAs show reduced EV secretion during LPS stimulation, and EVs isolated from SCT or SR knocked down cholangiocytes fail to induce inflammatory reactions in control large cholangiocytes. This study identifies cholangiocyte EV communication during LPS stimulation, and demonstrates that the SCT/SR axis may be important for this event.
Collapse
Affiliation(s)
- Keisaku Sato
- Research, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.,Department of Medicine, Texas A&M College of Medicine, Temple, TX, 76504, USA.,Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, TX, 76504, USA
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, TX, 76504, USA. .,Department of Medicine, Texas A&M College of Medicine, Temple, TX, 76504, USA. .,Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, TX, 76504, USA. .,Academic Research Integration, Baylor Scott & White Healthcare, Temple, TX, 76504, USA.
| | - Julie Venter
- Research, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.,Department of Medicine, Texas A&M College of Medicine, Temple, TX, 76504, USA.,Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, TX, 76504, USA
| | - Thao Giang
- Research, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.,Department of Medicine, Texas A&M College of Medicine, Temple, TX, 76504, USA.,Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, TX, 76504, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, TX, 76504, USA. .,Department of Medicine, Texas A&M College of Medicine, Temple, TX, 76504, USA. .,Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, TX, 76504, USA.
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, TX, 76504, USA. .,Department of Medicine, Texas A&M College of Medicine, Temple, TX, 76504, USA. .,Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, TX, 76504, USA.
| |
Collapse
|
131
|
Berardocco M, Radeghieri A, Busatto S, Gallorini M, Raggi C, Gissi C, D'Agnano I, Bergese P, Felsani A, Berardi AC. RNA-seq reveals distinctive RNA profiles of small extracellular vesicles from different human liver cancer cell lines. Oncotarget 2017; 8:82920-82939. [PMID: 29137313 PMCID: PMC5669939 DOI: 10.18632/oncotarget.20503] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/29/2017] [Indexed: 12/20/2022] Open
Abstract
Liver cancer (LC) is one of the most common cancers and represents the third highest cause of cancer-related deaths worldwide. Extracellular vesicle (EVs) cargoes, which are selectively enriched in RNA, offer great promise for the diagnosis, prognosis and treatment of LC. Our study analyzed the RNA cargoes of EVs derived from 4 liver-cancer cell lines: HuH7, Hep3B, HepG2 (hepato-cellular carcinoma) and HuH6 (hepatoblastoma), generating two different sets of sequencing libraries for each. One library was size-selected for small RNAs and the other targeted the whole transcriptome. Here are reported genome wide data of the expression level of coding and non-coding transcripts, microRNAs, isomiRs and snoRNAs providing the first comprehensive overview of the extracellular-vesicle RNA cargo released from LC cell lines. The EV-RNA expression profiles of the four liver cancer cell lines share a similar background, but cell-specific features clearly emerge showing the marked heterogeneity of the EV-cargo among the individual cell lines, evident both for the coding and non-coding RNA species.
Collapse
Affiliation(s)
- Martina Berardocco
- U.O.C. of Immunohaematology and Transfusion Medicine, Laboratory of Stem Cells, Spirito Santo Hospital, Pescara, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Busatto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Chiara Raggi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Clarissa Gissi
- U.O.C. of Immunohaematology and Transfusion Medicine, Laboratory of Stem Cells, Spirito Santo Hospital, Pescara, Italy
| | - Igea D'Agnano
- Institute of Cell Biology and Neurobiology, CNR, Monterotondo, Italy
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Armando Felsani
- Institute of Cell Biology and Neurobiology, CNR, Monterotondo, Italy.,Genomnia Srl, Bresso, Italy
| | - Anna C Berardi
- U.O.C. of Immunohaematology and Transfusion Medicine, Laboratory of Stem Cells, Spirito Santo Hospital, Pescara, Italy
| |
Collapse
|
132
|
Xie YX, Liao R, Pan L, Du CY. ERK pathway activation contributes to the tumor-promoting effects of hepatic stellate cells in hepatocellular carcinoma. Immunol Lett 2017; 188:116-123. [PMID: 28668554 DOI: 10.1016/j.imlet.2017.06.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/05/2017] [Accepted: 06/26/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Activated hepatic stellate cell (aHSC) play a critical role in hepatocellular carcinoma (HCC) progression crosstalking with cancer cell via various signaling pathways. The aim of our study is to explore the tumor-promoting effects of aHSCs on HCC via ERK pathway. METHODS α-SMA, p-ERK and p-JNK expression levels in tumoral and peritumoral tissues of HCC were assessed by immunohistochemical and western blotting. The protein and mRNA expression levels in human hepatoma cell treated with aHSC conditioned medium (CM) were determined by western blotting and real-time quantitative PCR, respectively. Cell migration and invasion abilities were assessed using transwell assays. The proliferation ability of HCC cells induced by aHSCs-CM was detected by CCK-8 assay and cell cycle analysis. RESULTS We found that aHSC number was positively correlated with p-ERK expression levels in tumoral tissues and aHSC-CM could time-dependently promote PCNA, p-ERK expression in HCC cells. Moreover, aHSC-CM enhanced HCC cells proliferation via ERK. Additionally, aHSC upregulated c-jun and cyclinD1 expression levels, accelerating the transition from G1 to the S phase of HCC cells, and this effect could be arrested by inhibiting ERK pathway. Furthermore, aHSC-CM promoted migration and invasion of HCC cells via ERK. Epithelial-mesenchymal transitions (EMT) phenomenon could be reversed by ERK suppression. CONCLUSION High expression of p-ERK and aHSCs may be associated with the aggressive behavior of HCC cells. Secretions from aHSCs could promote proliferation and EMT of HCC cells via ERK1/2/c-jun/cyclinD1 axis or ERK pathway.
Collapse
Affiliation(s)
- Yu-Xiao Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Rui Liao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Long Pan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cheng-You Du
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
133
|
Ge X, Wang Y, Nie J, Li Q, Tang L, Deng X, Wang F, Xu B, Wu X, Zhang X, You Q, Miao L. The diagnostic/prognostic potential and molecular functions of long non-coding RNAs in the exosomes derived from the bile of human cholangiocarcinoma. Oncotarget 2017; 8:69995-70005. [PMID: 29050258 PMCID: PMC5642533 DOI: 10.18632/oncotarget.19547] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignancy associated with unfavorable prognosis, and it’s difficult to diagnose and no effective treatments are available. Long non-coding RNAs (lncRNAs) play important roles in tumorigenesis and metastasis. Intact lncRNAs from exosomes have sparked much interest as potential biomarker for the non-invasive analysis of disease. Here, via exosome sequencing on lncRNAs, GO analysis, KEGG pathway and co-expression analysis, receiver operating characteristic curve and survival analyses, we found that, compared with control group, lncRNAs of ENST00000588480.1 and ENST00000517758.1 showed significantly increased expressions in CCA group. Moreover, area under the curve (AUC) was increased to 0.709 when combined the two lncRNAs, they had a sensitivity and specificity of 82.9% and 58.9% respectively. Further, the higher levels of the two lncRNAs showed a significantly increasing trend with the advancement of cancer TNM stages, and prognosticated a poor survival. In addition, KEGG pathway analysis showed that the most significant difference term was “p53 signaling pathway” (KEGG ID: hsa04115, p: 0.001). The altered lncRNAs and their target genes were included to reconstruct a co-expression network. These altered lncRNAs were mainly related to cellular processes, environmental information processing and organismal systems, etc. Collectively, our findings provided the potential roles of lncRNAs of ENST00000588480.1 and ENST00000517758.1 in CCA, and implicated these lncRNAs as potential diagnostic and therapeutic targets for CCA.
Collapse
Affiliation(s)
- Xianxiu Ge
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Youli Wang
- Department of Clinical Laboratory, Nanjing First Hospital, Nanjing, China
| | - Junjie Nie
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Quanpeng Li
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lingyu Tang
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xueting Deng
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei Wang
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Boming Xu
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaochao Wu
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuhua Zhang
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiang You
- Department of Biotherapy, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Miao
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
134
|
Cai Y, Xu MJ, Koritzinsky EH, Zhou Z, Wang W, Cao H, Yuen PS, Ross RA, Star RA, Liangpunsakul S, Gao B. Mitochondrial DNA-enriched microparticles promote acute-on-chronic alcoholic neutrophilia and hepatotoxicity. JCI Insight 2017; 2:92634. [PMID: 28724791 DOI: 10.1172/jci.insight.92634] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 06/13/2017] [Indexed: 12/16/2022] Open
Abstract
Over the last several years, one of the major advances in the field of alcoholic liver disease research was the discovery that binge alcohol consumption induced neutrophilia and hepatic neutrophil infiltration in chronically ethanol-fed mice and human subjects with excessive alcohol use (EAU); however, the underlying mechanisms remain obscure. Here, we demonstrated that chronic EAU patients with a history of recent excessive drinking (EAU + RD) had higher serum levels of mitochondrial DNA (mtDNA)-enriched microparticles (MPs) than EAU without recent drinking (EAU - RD) and healthy controls, which correlated positively with circulating neutrophils. Similarly, mice with chronic-plus-binge (E10d + 1B) ethanol feeding also had markedly elevated serum levels of mtDNA-enriched MPs, with activation of hepatic ER stress and inflammatory responses. Inhibition of ER stress by gene KO or inhibitors attenuated ethanol-induced elevation of mtDNA-enriched MPs, neutrophilia, and liver injury. The data from the study of hepatocyte-specific deletion of the protein kinase RNA-like ER kinase (Perk) gene in mice and of cultured hepatocytes demonstrated that hepatocytes were the main source of mtDNA-enriched MPs after ethanol feeding. Finally, administration of mtDNA-enriched MPs isolated from E10d+1B-fed mice caused neutrophilia in mice. In conclusion, E10d + 1B ethanol consumption activates hepatic ER stress-dependent mtDNA-enriched MP release, leading to neutrophilia and liver injury.
Collapse
Affiliation(s)
- Yan Cai
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA)
| | - Ming-Jiang Xu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA)
| | - Erik H Koritzinsky
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Zhou Zhou
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA)
| | - Wei Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA)
| | - Haixia Cao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA)
| | - Peter St Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Ruth A Ross
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA)
| |
Collapse
|
135
|
Taleb RSZ, Moez P, Younan D, Eisenacher M, Tenbusch M, Sitek B, Bracht T. Quantitative proteome analysis of plasma microparticles for the characterization of HCV-induced hepatic cirrhosis and hepatocellular carcinoma. Proteomics Clin Appl 2017. [PMID: 28626882 DOI: 10.1002/prca.201700014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is the most common primary malignant liver tumor and a leading cause of cancer-related deaths worldwide. Cirrhosis induced by hepatitis-C virus (HCV) infection is the most critical risk factor for HCC. However, the mechanism of HCV-induced carcinogenesis is not fully understood. Plasma microparticles (PMP) contribute to numerous physiological and pathological processes and contain proteins whose composition correlates to the respective pathophysiological conditions. EXPERIMENTAL DESIGN We analyzed PMP from 22 HCV-induced cirrhosis patients, 16 HCV-positive HCC patients with underlying cirrhosis and 18 healthy controls. PMP were isolated using ultracentrifugation and analyzed via label-free LC-MS/MS. RESULTS We identified 840 protein groups and quantified 507 proteins. 159 proteins were found differentially abundant between the three experimental groups. PMP in both disease entities displayed remarkable differences in the proteome composition compared to healthy controls. Conversely, the proteome difference between both diseases was minimal. GO analysis revealed that PMP isolated from both diseases were significantly enriched in proteins involved in complement activation, while endopeptidase activity was downregulated exclusively in HCC patients. CONCLUSION This study reports for the first time a quantitative proteome analysis for PMP from patients with HCV-induced cirrhosis and HCC. Data are available via ProteomeXchange with identifier PXD005777.
Collapse
Affiliation(s)
- Raghda Saad Zaghloul Taleb
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany.,Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Pacint Moez
- Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Doreen Younan
- Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Matthias Tenbusch
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Thilo Bracht
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany
| |
Collapse
|
136
|
Zhang J, Lu S, Zhou Y, Meng K, Chen Z, Cui Y, Shi Y, Wang T, He QY. Motile hepatocellular carcinoma cells preferentially secret sugar metabolism regulatory proteins via exosomes. Proteomics 2017; 17. [PMID: 28590090 DOI: 10.1002/pmic.201700103] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/14/2017] [Accepted: 05/29/2017] [Indexed: 01/29/2023]
Abstract
Exosomes are deliverers of critically functional proteins, capable of transforming target cells in numerous cancers, including hepatocellular carcinoma (HCC). We hypothesize that the motility of HCC cells can be featured by comparative proteome of exosomes. Hence, we performed the super-SILAC-based MS analysis on the exosomes secreted by three human HCC cell lines, including the non-motile Hep3B cell, and the motile 97H and LM3 cells. More than 1400 exosomal proteins were confidently quantified in each MS analysis with highly biological reproducibility. We justified that 469 and 443 exosomal proteins represented differentially expressed proteins (DEPs) in the 97H/Hep3B and LM3/Hep3B comparisons, respectively. These DEPs focused on sugar metabolism-centric canonical pathways per ingenuity pathway analysis, which was consistent with the gene ontology analysis on biological process enrichment. These pathways included glycolysis I, gluconeogenesis I and pentose phosphate pathways; and the DEPs enriched in these pathways could form a tightly connected network. By analyzing the relative abundance of proteins and translating mRNAs, we found significantly positive correlation between exosomes and cells. The involved exosomal proteins were again focusing on sugar metabolism. In conclusion, motile HCC cells tend to preferentially export more sugar metabolism-associated proteins via exosomes that differentiate them from non-motile HCC cells.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Shaohua Lu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Ye Zhou
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Kun Meng
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Zhipeng Chen
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Yizhi Cui
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Yunfeng Shi
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Tong Wang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| |
Collapse
|
137
|
New insight into inter-organ crosstalk contributing to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Protein Cell 2017. [PMID: 28643267 PMCID: PMC5818366 DOI: 10.1007/s13238-017-0436-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver dysfunction and a significant global health problem with substantial rise in prevalence over the last decades. It is becoming increasingly clear that NALFD is not only predominantly a hepatic manifestation of metabolic syndrome, but also involves extra-hepatic organs and regulatory pathways. Therapeutic options are limited for the treatment of NAFLD. Accordingly, a better understanding of the pathogenesis of NAFLD is critical for gaining new insight into the regulatory network of NAFLD and for identifying new targets for the prevention and treatment of NAFLD. In this review, we emphasize on the current understanding of the inter-organ crosstalk between the liver and peripheral organs that contributing to the pathogenesis of NAFLD.
Collapse
|
138
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a dominant cause of chronic liver disease, but the exact mechanism of progression from simple steatosis to nonalcoholic steatohepatitis (NASH) remains unknown. Here, we investigated the role of exosomes in NAFLD progression. Exosomes were isolated from a human hepatoma cell line treated with palmitic acid (PA) and their miRNA profiles examined by microarray. The human hepatic stellate cell (HSC) line (LX-2) was then treated with exosome isolated from hepatocytes. Compared with controls, PA-treated hepatocytes displayed significantly increased CD36 and exosome production. The microarray analysis showed there to be distinctive miRNA expression patterns between exosomes from vehicle- and PA-treated hepatocytes. When LX-2 cells were cultured with exosomes from PA-treated hepatocytes, the expression of genes related to the development of fibrosis were significantly amplified compared to those treated with exosomes from vehicle-treated hepatocytes. In conclusion, PA treatment enhanced the production of exosomes in these hepatocytes and changed their exosomal miRNA profile. Moreover, exosomes derived from PA-treated hepatocytes caused an increase in the expression levels of fibrotic genes in HSCs. Therefore, exosomes may have important roles in the crosstalk between hepatocytes and HSCs in the progression from simple steatosis to NASH.
Collapse
|
139
|
Wang S, Wang JQ, Lv XW. Exosomal miRNAs as biomarkers in the diagnosis of liver disease. Biomark Med 2017; 11:491-501. [PMID: 28598214 DOI: 10.2217/bmm-2017-0011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Liver disease is a primary cause of liver-related morbidity and mortality worldwide. Currently, histological examination is the gold standard for diagnosis and classification of liver disease; however, due to its several drawbacks, including the risk of complications and sampling variability, noninvasive diagnostic options are favorable. Exosomal miRNAs have recently been considered as an important source of medical biomarkers due to being widely distributed in body fluids. This review summarizes existing concepts related to the origin, mode of transportation and possible functions of exosomal miRNAs, and recent findings on the role of exosomal miRNAs in liver diseases and development of exosomal miRNA-based diagnostic biomarkers in the primary forms of liver diseases.
Collapse
Affiliation(s)
- Sheng Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Jian-Qing Wang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Xiong-Wen Lv
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.,Institute for Liver Disease of Anhui Medical University, Anhui Medical University, Hefei 230032, China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
140
|
Lou G, Yang Y, Liu F, Ye B, Chen Z, Zheng M, Liu Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J Cell Mol Med 2017; 21:2963-2973. [PMID: 28544786 PMCID: PMC5661245 DOI: 10.1111/jcmm.13208] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/11/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation alone may be insufficient for treatment of liver fibrosis because of complicated histopathological changes in the liver. Given that miR‐122 plays an essential role in liver fibrosis by negatively regulating the proliferation and transactivation of hepatic stellate cells (HSCs), this study investigated whether miR‐122 modification can improve the therapeutic efficacy of adipose tissue‐derived MSCs in treating liver fibrosis. MiR‐122‐modified AMSCs (AMSC‐122) were constructed through lentivirus‐mediated transfer of pre‐miR‐122. MiR‐122‐modified AMSCs expressed high level of miR‐122, while they retained their phenotype and differentiation potential as naïve AMSCs. AMSC‐122 more effectively suppressed the proliferation of and collagen maturation in HSCs than scramble miRNA‐modified AMSCs. In addition, AMSC‐derived exosomes mediated the miR‐122 communication between AMSCs and HSCs, further affecting the expression levels of miR‐122 target genes, such as insulin‐like growth factor receptor 1 (IGF1R), Cyclin G(1) (CCNG1) and prolyl‐4‐hydroxylase α1 (P4HA1), which are involved in proliferation of and collagen maturation in HSCs. Moreover, miR‐122 modification enhanced the therapeutic efficacy of AMSCs in the treatment of carbon tetrachloride (CCl4)‐induced liver fibrosis by suppressing the activation of HSCs and alleviating collagen deposition. Results demonstrate that miR‐122 modification improves the therapeutic efficacy of AMSCs through exosome‐mediated miR‐122 communication; thus, miR‐122 modification is a new potential strategy for treatment of liver fibrosis.
Collapse
Affiliation(s)
- Guohua Lou
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Yang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feifei Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingjue Ye
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Chen
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min Zheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanning Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
141
|
Chapuy-Regaud S, Dubois M, Plisson-Chastang C, Bonnefois T, Lhomme S, Bertrand-Michel J, You B, Simoneau S, Gleizes PE, Flan B, Abravanel F, Izopet J. Characterization of the lipid envelope of exosome encapsulated HEV particles protected from the immune response. Biochimie 2017; 141:70-79. [PMID: 28483690 DOI: 10.1016/j.biochi.2017.05.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/03/2017] [Indexed: 02/08/2023]
Abstract
The hepatitis E virus (HEV) is the most common cause of acute hepatitis worldwide. Although HEV is a small, naked RNA virus, HEV particles become associated with lipids in the blood of infected patients and in the supernatant of culture systems. The egress of these particles from cells implies the exocytosis pathway but the question of the role of the resulting HEV RNA containing exosomes and the nature of the lipids they contain has not been fully addressed. We determined the lipid proportions of exosomes from uninfected and HEV-infected cells and their role in HEV spreading. We cultured a suitable HEV strain on HepG2/C3A cells and analyzed the population of exosomes containing HEV RNA using lipidomics methods and electron microscopy. We also quantified HEV infectivity using an infectivity endpoint method based on HEV RNA quantification to calculate the tissue culture infectious dose 50. Exosomes produced by HEV-infected HepG2/C3A cells contained encapsidated HEV RNA. These HEV RNA-containing exosomes were infectious but ten times less than stools. HEV from stools, but not exosome-associated HEV from culture supernatant, was neutralized by anti-HEV antibodies in a dose-dependent manner. HEV infection did not influence the morphology or lipid proportions of the bulk of exosomes. These exosomes contained significantly more cholesterol, phosphatidylserine, sphingomyelin and ceramides than the parent cells, but less phosphoinositides and polyunsaturated fatty acids. Exosomes play a major role in HEV egress but HEV infection does not modify the characteristics of the bulk of exosomes produced by infected cells. PS and cholesterol enriched in these vesicles could then be critical for HEV entry. HEV particles in exosomes are protected from the immune response which could lead to the wide circulation of HEV in its host.
Collapse
Affiliation(s)
- Sabine Chapuy-Regaud
- INSERM, UMR1043, Toulouse, France; Department of Virology, CHU Purpan, Toulouse, France; Toulouse University, Toulouse, France.
| | - Martine Dubois
- INSERM, UMR1043, Toulouse, France; Department of Virology, CHU Purpan, Toulouse, France
| | | | - Tiffany Bonnefois
- INSERM, UMR1043, Toulouse, France; Department of Virology, CHU Purpan, Toulouse, France
| | - Sébastien Lhomme
- INSERM, UMR1043, Toulouse, France; Department of Virology, CHU Purpan, Toulouse, France; Toulouse University, Toulouse, France
| | - Justine Bertrand-Michel
- Toulouse University, Toulouse, France; MetaToul-Lipidomic Core Facility, INSERM, UMR1048, Toulouse, France
| | - Bruno You
- LFB, Laboratoire Français du Fractionnement et des Biotechnologies, Courtaboeuf, France
| | - Steve Simoneau
- LFB, Laboratoire Français du Fractionnement et des Biotechnologies, Courtaboeuf, France
| | | | - Benoît Flan
- LFB, Laboratoire Français du Fractionnement et des Biotechnologies, Courtaboeuf, France
| | - Florence Abravanel
- INSERM, UMR1043, Toulouse, France; Department of Virology, CHU Purpan, Toulouse, France; Toulouse University, Toulouse, France
| | - Jacques Izopet
- INSERM, UMR1043, Toulouse, France; Department of Virology, CHU Purpan, Toulouse, France; Toulouse University, Toulouse, France
| |
Collapse
|
142
|
Loosen SH, Schueller F, Trautwein C, Roy S, Roderburg C. Role of circulating microRNAs in liver diseases. World J Hepatol 2017; 9:586-594. [PMID: 28515844 PMCID: PMC5411953 DOI: 10.4254/wjh.v9.i12.586] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are small RNAs regulate gene expression by inhibiting the turnover of their target mRNAs. In the last years, it became apparent that miRNAs are released into the circulation and circulating miRNAs emerged as a new class of biomarkers for various diseases. In this review we summarize available data on the role of circulating miRNAs in the context of acute and chronic liver diseases including hepatocellular and cholangiocellular carcinoma. Data from animal models are compared to human data and current challenges in the field of miRNAs research are discussed.
Collapse
|
143
|
Exosome-Mediated Intercellular Communication between Hepatitis C Virus-Infected Hepatocytes and Hepatic Stellate Cells. J Virol 2017; 91:JVI.02225-16. [PMID: 28077652 DOI: 10.1128/jvi.02225-16] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/04/2017] [Indexed: 12/15/2022] Open
Abstract
Fibrogenic pathways in the liver are principally regulated by activation of hepatic stellate cells (HSC). Fibrosis is associated with chronic hepatitis C virus (HCV) infection, although the mechanism is poorly understood. HSC comprise the major population of nonparenchymal cells in the liver. Since HCV does not replicate in HSC, we hypothesized that exosomes secreted from HCV-infected hepatocytes activate HSC. Primary or immortalized human hepatic stellate (LX2) cells were exposed to exosomes derived from HCV-infected hepatocytes (HCV-exo), and the expression of fibrosis-related genes was examined. Our results demonstrated that HCV-exo internalized to HSC and increased the expression of profibrotic markers. Further analysis suggested that HCV-exo carry miR-19a and target SOCS3 in HSC, which in turn activates the STAT3-mediated transforming growth factor β (TGF-β) signaling pathway and enhances fibrosis marker genes. The higher expression of miR-19a in exosomes was also observed from HCV-infected hepatocytes and in sera of chronic HCV patients with fibrosis compared to healthy volunteers and non-HCV-related liver disease patients with fibrosis. Together, our results demonstrated that miR-19a carried through the exosomes from HCV-infected hepatocytes activates HSC by modulating the SOCS-STAT3 axis. Our results implicated a novel mechanism of exosome-mediated intercellular communication in the activation of HSC for liver fibrosis in HCV infection.IMPORTANCE HCV-associated liver fibrosis is a critical step for end-stage liver disease progression. However, the molecular mechanisms for hepatic stellate-cell activation by HCV-infected hepatocytes are underexplored. Here, we provide a role for miR-19a carried through the exosomes in intercellular communication between HCV-infected hepatocytes and HSC in fibrogenic activation. Furthermore, we demonstrate the role of exosomal miR-19a in activation of the STAT3-TGF-β pathway in HSC. This study contributes to the understanding of intercellular communication in the pathogenesis of liver disease during HCV infection.
Collapse
|
144
|
Gradilone SA. Extracellular vesicles as therapeutic carriers of microRNAs for cholangiocarcinoma. Hepatology 2017; 65:404-406. [PMID: 28012192 PMCID: PMC5507678 DOI: 10.1002/hep.28925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/19/2016] [Accepted: 10/22/2016] [Indexed: 12/20/2022]
|
145
|
Huang-Doran I, Zhang CY, Vidal-Puig A. Extracellular Vesicles: Novel Mediators of Cell Communication In Metabolic Disease. Trends Endocrinol Metab 2017; 28:3-18. [PMID: 27810172 DOI: 10.1016/j.tem.2016.10.003] [Citation(s) in RCA: 274] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/16/2016] [Accepted: 10/03/2016] [Indexed: 12/27/2022]
Abstract
Metabolic homeostasis emerges from the complex, multidirectional crosstalk between key metabolic tissues including adipose tissue, liver, and skeletal muscle. This crosstalk, traditionally mediated by hormones and metabolites, becomes dysregulated in human diseases such as obesity and diabetes. Extracellular vesicles (EVs; including exosomes) are circulating, cell-derived nanoparticles containing proteins and nucleic acids that interact with and modify local and distant cellular targets. Accumulating evidence, reviewed herein, supports a role for extracellular vesicles in obesity-associated metabolic disturbance, particularly the local and systemic inflammation characteristic of adipose and hepatic stress. As the practical and conceptual challenges facing the field are tackled, this emerging and versatile mode of intercellular communication may afford valuable insights and therapeutic opportunities in combatting these major threats to modern human health.
Collapse
Affiliation(s)
- Isabel Huang-Doran
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK.
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute for Life Sciences, Nanjing University, 163 Xianlin Road, Nanjing, Jiangsu, 210046 China
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
146
|
Hirsova P, Ibrahim SH, Verma VK, Morton LA, Shah VH, LaRusso NF, Gores GJ, Malhi H. Extracellular vesicles in liver pathobiology: Small particles with big impact. Hepatology 2016; 64:2219-2233. [PMID: 27628960 PMCID: PMC5115968 DOI: 10.1002/hep.28814] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/26/2016] [Accepted: 08/10/2016] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are nanometer-sized, membrane-bound vesicles released by cells into the extracellular milieu. EVs are now recognized to play a critical role in cell-to-cell communication. EVs contain important cargo in the form of proteins, lipids, and nucleic acids and serve as vectors for delivering this cargo from donor to acceptor or target cell. EVs are released under both physiologic and pathologic conditions, including liver diseases, and exert a wide range of effects on target cells. This review provides an overview on EV biogenesis, secretion, cargo, and target cell interactions in the context of select liver diseases. Specifically, the diverse roles of EVs in nonalcoholic steatohepatitis, alcoholic liver disease, viral hepatitis, cholangiopathies, and hepatobiliary malignancies are emphasized. Liver diseases often result in an increased release of EVs and/or in different cargo sorting into these EVs. Either of these alterations can drive disease pathogenesis. Given this fact, EVs represent a potential target for therapeutic intervention in liver disorders. Because altered EV composition may reflect the underlying disease condition, circulating EVs can be exploited for diagnostic and prognostic purposes as a liquid biopsy. Furthermore, ex vivo modified or synthesized EVs can be engineered as therapeutic nano-shuttles. Finally, we highlight areas that merit further investigation relevant to understanding how EVs regulate liver disease pathogenesis. (Hepatology 2016;64:2219-2233).
Collapse
Affiliation(s)
- Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Samar H. Ibrahim
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Vikas K. Verma
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Leslie A. Morton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
147
|
Benhamouche-Trouillet S, Postic C. Emerging role of miR-21 in non-alcoholic fatty liver disease. Gut 2016; 65:1781-1783. [PMID: 27436271 DOI: 10.1136/gutjnl-2015-310044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 06/23/2016] [Accepted: 06/26/2016] [Indexed: 12/08/2022]
Affiliation(s)
| | - Catherine Postic
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
148
|
Akrap I, Thavamani A, Nordheim A. Vps4A-mediated tumor suppression upon exosome modulation? ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:180. [PMID: 27275493 DOI: 10.21037/atm.2016.04.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Ivana Akrap
- Molecular Biology Unit, Interfaculty Institute of Cell Biology, Tübingen University, Tübingen, Germany
| | - Abhishek Thavamani
- Molecular Biology Unit, Interfaculty Institute of Cell Biology, Tübingen University, Tübingen, Germany
| | - Alfred Nordheim
- Molecular Biology Unit, Interfaculty Institute of Cell Biology, Tübingen University, Tübingen, Germany
| |
Collapse
|
149
|
Exosomes: multitask cargo carriers modulating innate immunity to viruses. Cell Mol Immunol 2016; 14:476-477. [PMID: 27264688 DOI: 10.1038/cmi.2016.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/05/2016] [Accepted: 05/05/2016] [Indexed: 11/08/2022] Open
|