1
|
Felippe TVD, Toro DM, de Carvalho JCS, Nobre-Azevedo P, Rodrigues LFM, Oliveira BTM, da Silva-Neto PV, Vilela AFL, Almeida F, Faccioli LH, Sorgi CA. High-resolution targeted mass spectrometry for comprehensive quantification of sphingolipids: clinical applications and characterization of extracellular vesicles. Anal Biochem 2025; 698:115732. [PMID: 39622401 DOI: 10.1016/j.ab.2024.115732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Sphingolipids (SL), a class of membrane lipids, play important roles in numerous biological processes. Their significant structural diversity poses challenges for accurate quantification. To address this, liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) has emerged as a powerful tool for sphingolipidomics, capable of profiling these lipids comprehensively. In this study, we utilized LC-MS/MS with high-resolution mass spectrometry (MRMHR) to develop a targeted method for the identification and quantification of various SL species. This method, based on validated parameters such as precursor/fragment ions (m/z) and retention time, demonstrated high sensitivity and accuracy, successfully identifying SL species across 12 distinct classes. Its open-panel design also facilitates the analysis of new SL-species targets. Notably, using this approach, we identified 40 SL species in plasma samples from COVID-19 patients, and we determined the influence of matrix metalloproteinase-3 (MMP-3) expression on the positive downstream of SL metabolism. Beyond plasma analysis, this method has potential applications in other biomedical contexts, such as extracellular vesicles (EVs), describing the cargo of sphingosine-1-phosphate (S1P) on macrophage-derived EVs. The establishment of this targeted workflow enabling precise quantification of a wide range of SL species, holds promise for identifying novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Thiago V D Felippe
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada - PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas - UFAM, Manaus, 69080-900, AM, Brazil
| | - Jonatan C S de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil
| | - Pedro Nobre-Azevedo
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil
| | - Luiz F M Rodrigues
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil
| | - Bianca T M Oliveira
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil
| | - Pedro V da Silva-Neto
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada - PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas - UFAM, Manaus, 69080-900, AM, Brazil
| | - Adriana F L Vilela
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil
| | - Fausto Almeida
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil; Centro de Excelência em Quantificação e Identificação de Lipídios (CEQIL), Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil
| | - Carlos A Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada - PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas - UFAM, Manaus, 69080-900, AM, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil; Centro de Excelência em Quantificação e Identificação de Lipídios (CEQIL), Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil.
| |
Collapse
|
2
|
Ismail M, Fadul MM, Taha R, Siddig O, Elhafiz M, Yousef BA, Jiang Z, Zhang L, Sun L. Dynamic role of exosomal long non-coding RNA in liver diseases: pathogenesis and diagnostic aspects. Hepatol Int 2024; 18:1715-1730. [PMID: 39306594 DOI: 10.1007/s12072-024-10722-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND Liver disease has emerged as a significant health concern, characterized by high rates of morbidity and mortality. Circulating exosomes have garnered attention as important mediators of intercellular communication, harboring protein and stable mRNAs, microRNAs, and long non-coding RNAs (lncRNA). This review highlights the involvement of exosomal lncRNA in the pathogenesis and diagnosis of various liver diseases. Notably, exosomal lncRNAs exhibit therapeutic potential as targets for conditions including hepatic carcinoma, hepatic fibrosis, and hepatic viral infections. METHOD An online screening process was employed to identify studies investigating the association between exosomal lncRNA and various liver diseases. RESULT Our study revealed a diverse array of lncRNAs carried by exosomes, including H19, Linc-ROR, VLDLR, MALAT1, DANCR, HEIH, ENSG00000248932.1, ENST00000457302.2, ZSCAN16-AS1, and others, exhibiting varied levels across different liver diseases compared to normal liver tissue. These exosomal-derived lncRNAs are increasingly recognized as pivotal biomarkers for diagnosing and prognosticating liver diseases, supported by emerging evidence. However, the precise mechanisms underlying the involvement of certain exosomal lncRNAs remain incompletely understood. Furthermore, the combined analysis of serum exosomes using ENSG00000258332.1, LINC00635, and serum AFP may serve as novel and valuable biomarker for HCC. Clinically, exosomal ATB expression is upregulated in HCC, while exosomal HEIH and RP11-513I15.6 have shown potential for distinguishing HCC related to HCV infection. CONCLUSION The lack of reliable biomarkers for liver diseases, coupled with the high specificity and sensitivity of exosomal lncRNA and its non-invasive detection, promotes exploring their role in pathogenesis and biomarker for diagnosis, prognosis, and response to treatment liver diseases.
Collapse
Affiliation(s)
- Mohammed Ismail
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacology, Faculty of Medicine and Health Science, Dongola University, Dongola, Sudan
| | - Missaa M Fadul
- Department of Pharmacology, Faculty of Medicine and Health Science, Dongola University, Dongola, Sudan
| | - Reham Taha
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Orwa Siddig
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Muhanad Elhafiz
- Department of Pharmacology, Faculty of Pharmacy, Omdurman Islamic University, Khartoum, Sudan
| | - Bashir A Yousef
- Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Zhenzhou Jiang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyong Zhang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Lixin Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
3
|
Lucena MI, Villanueva-Paz M, Alvarez-Alvarez I, Aithal GP, Björnsson ES, Cakan-Akdogan G, Cubero FJ, Esteves F, Falcon-Perez JM, Fromenty B, Garcia-Ruiz C, Grove JI, Konu O, Kranendonk M, Kullak-Ublick GA, Miranda JP, Remesal-Doblado A, Sancho-Bru P, Nelson L, Andrade RJ, Daly AK, Fernandez-Checa JC. Roadmap to DILI research in Europe. A proposal from COST action ProEuroDILINet. Pharmacol Res 2024; 200:107046. [PMID: 38159783 DOI: 10.1016/j.phrs.2023.107046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
In the current article the aims for a constructive way forward in Drug-Induced Liver Injury (DILI) are to highlight the most important priorities in research and clinical science, therefore supporting a more informed, focused, and better funded future for European DILI research. This Roadmap aims to identify key challenges, define a shared vision across all stakeholders for the opportunities to overcome these challenges and propose a high-quality research program to achieve progress on the prediction, prevention, diagnosis and management of this condition and impact on healthcare practice in the field of DILI. This will involve 1. Creation of a database encompassing optimised case report form for prospectively identified DILI cases with well-characterised controls with competing diagnoses, biological samples, and imaging data; 2. Establishing of preclinical models to improve the assessment and prediction of hepatotoxicity in humans to guide future drug safety testing; 3. Emphasis on implementation science and 4. Enhanced collaboration between drug-developers, clinicians and regulatory scientists. This proposed operational framework will advance DILI research and may bring together basic, applied, translational and clinical research in DILI.
Collapse
Affiliation(s)
- M I Lucena
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Plataforma de Investigación Clínica y Ensayos Clínicos UICEC-IBIMA, Plataforma ISCIII de Investigación Clínica, Madrid, Spain.
| | - M Villanueva-Paz
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - I Alvarez-Alvarez
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - G P Aithal
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom; NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, United Kingdom
| | - E S Björnsson
- Faculty of Medicine, University of Iceland, Department of Gastroenterology and Hepatology, Landspitali University Hospital, Reykjavik, Iceland
| | - G Cakan-Akdogan
- Izmir Biomedicine and Genome Center, Izmir, Turkey. Department of Medical Biology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | - F J Cubero
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - F Esteves
- Center for Toxicogenomics and Human Health (ToxOmics), NMS | FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - J M Falcon-Perez
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain. IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia 48009, Spain
| | - B Fromenty
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1317, F-35000 Rennes, France
| | - C Garcia-Ruiz
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. University of Barcelona, Barcelona, Spain; Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain
| | - J I Grove
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom; NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, United Kingdom
| | - O Konu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey; Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - M Kranendonk
- Center for Toxicogenomics and Human Health (ToxOmics), NMS | FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - G A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; CMO & Patient Safety, Global Drug Development, Novartis Pharma, Basel, Switzerland
| | - J P Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - A Remesal-Doblado
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain
| | - P Sancho-Bru
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. University of Barcelona, Barcelona, Spain
| | - L Nelson
- Institute for Bioengineering, School of Engineering, Faraday Building, The University of Edinburgh, Scotland, UK
| | - R J Andrade
- Servicios de Aparato Digestivo y Farmacología Clínica, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Universidad de Málaga, Málaga, Spain; Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - A K Daly
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - J C Fernandez-Checa
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain. University of Barcelona, Barcelona, Spain; Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, Barcelona, Spain; Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
4
|
Andújar-Vera F, Alés-Palmer ML, Muñoz-de-Rueda P, Iglesias-Baena I, Ocete-Hita E. Metabolomic Analysis of Pediatric Patients with Idiosyncratic Drug-Induced Liver Injury According to the Updated RUCAM. Int J Mol Sci 2023; 24:13562. [PMID: 37686369 PMCID: PMC10487599 DOI: 10.3390/ijms241713562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/20/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Hepatotoxicity, a common adverse drug effect, has been extensively studied in adult patients. However, it is equally important to investigate this condition in pediatric patients to develop personalized treatment strategies for children. This study aimed to identify plasma biomarkers that characterize hepatotoxicity in pediatric patients through an observational case-control study. Metabolomic analysis was conducted on 55 pediatric patients with xenobiotic liver toxicity and 88 healthy controls. The results revealed clear differences between the two groups. Several metabolites, including hydroxydecanoylcarnitine, octanoylcarnitine, lysophosphatidylcholine, glycocholic acid, and taurocholic acid, were identified as potential biomarkers (area under the curve: 0.817; 95% confidence interval: 0.696-0.913). Pathway analysis indicated involvement of primary bile acid biosynthesis and the metabolism of taurine and hypotaurine (p < 0.05). The findings from untargeted metabolomic analysis demonstrated an increase in bile acids in children with hepatotoxicity. The accumulation of cytotoxic bile acids should be further investigated to elucidate the role of these metabolites in drug-induced liver injury.
Collapse
Affiliation(s)
| | - María Luisa Alés-Palmer
- Department of Pediatrics, University of Granada, 18016 Granada, Spain;
- Department of Pediatrics, “Virgen de las Nieves” University Hospital, 18014 Granada, Spain
| | - Paloma Muñoz-de-Rueda
- Research Support Unit, Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain;
| | | | - Esther Ocete-Hita
- Department of Pediatrics, University of Granada, 18016 Granada, Spain;
- Department of Pediatrics, “Virgen de las Nieves” University Hospital, 18014 Granada, Spain
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
5
|
Gagliardi A, Bajraktari-Sylejmani G, Barocelli E, Weiss J, Rigalli JP. Extracellular Vesicles as Surrogates for Drug Metabolism and Clearance: Promise vs. Reality. Life (Basel) 2023; 13:1745. [PMID: 37629602 PMCID: PMC10455864 DOI: 10.3390/life13081745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Drug-metabolizing enzymes (DMEs) and transporters play a major role in drug efficacy and safety. They are regulated at multiple levels and by multiple factors. Estimating their expression and activity could contribute to predicting drug pharmacokinetics and their regulation by drugs or pathophysiological situations. Determining the expression of these proteins in the liver, intestine, and kidney requires the collection of biopsy specimens. Instead, the isolation of extracellular vesicles (EVs), which are nanovesicles released by most cells and present in biological fluids, could deliver this information in a less invasive way. In this article, we review the use of EVs as surrogates for the expression and activity of DMEs, uptake, and efflux transporters. Preliminary evidence has been provided for a correlation between the expression of some enzymes and transporters in EVs and the tissue of origin. In some cases, data obtained in EVs reflect the induction of phase I-DMEs in the tissues. Further studies are required to elucidate to what extent the regulation of other DMEs and transporters in the tissues reflects in the EV cargo. If an association between tissues and their EVs is firmly established, EVs may represent a significant advancement toward precision therapy based on the biotransformation and excretion capacity of each individual.
Collapse
Affiliation(s)
- Anna Gagliardi
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Gzona Bajraktari-Sylejmani
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Elisabetta Barocelli
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Juan Pablo Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
6
|
Jiang X, Wu S, Hu C. A narrative review of the role of exosomes and caveolin-1 in liver diseases and cancer. Int Immunopharmacol 2023; 120:110284. [PMID: 37196562 DOI: 10.1016/j.intimp.2023.110284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/16/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023]
Abstract
Exosomes are nanoscale (40-100 nm) vesicles secreted by different types of cells and have attracted extensive interest in recent years because of their unique role in disease development. It can carry related goods, such as lipids, proteins, and nucleic acids, to mediate intercellular communication. This review summarizes exosome biogenesis, release, uptake, and their role in mediating the development of liver diseases and cancer, such as viral hepatitis, drug-induced liver injury, alcohol-related liver disease, non-alcoholic fatty liver disease, hepatocellular carcinoma, and other tumors. Meanwhile, a fossa structural protein, caveolin-1(CAV-1), has also been proposed to be involved in the development of various diseases, especially liver diseases and tumors. In this review, we discuss the role of CAV-1 in liver diseases and different tumor stages (inhibition of early growth and promotion of late metastasis) and the underlying mechanisms by which CAV-1 regulates the process. In addition, CAV-1 has also been found to be a secreted protein that can be released directly through the exosome pathway or change the cargo composition of the exosomes, thus contributing to enhancing the metastasis and invasion of cancer cells during the late stage of tumor development. In conclusion, the role of CAV-1 and exosomes in disease development and the association between them remains to be one challenging uncharted area.
Collapse
Affiliation(s)
- Xiangfu Jiang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui medical university, Hefei 230032, China; Key Laboratory of anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Shuai Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui medical university, Hefei 230032, China; Key Laboratory of anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Chengmu Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui medical university, Hefei 230032, China; Key Laboratory of anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
7
|
Tamasi V, Németh K, Csala M. Role of Extracellular Vesicles in Liver Diseases. Life (Basel) 2023; 13:life13051117. [PMID: 37240762 DOI: 10.3390/life13051117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membrane structures that are formed by budding from the plasma membrane or originate from the endosomal system. These microparticles (100 nm-100 µm) or nanoparticles (>100 nm) can transport complex cargos to other cells and, thus, provide communication and intercellular regulation. Various cells, such as hepatocytes, liver sinusoidal endothelial cells (LSECs) or hepatic stellate cells (HSCs), secrete and take up EVs in the healthy liver, and the amount, size and content of these vesicles are markedly altered under pathophysiological conditions. A comprehensive knowledge of the modified EV-related processes is very important, as they are of great value as biomarkers or therapeutic targets. In this review, we summarize the latest knowledge on hepatic EVs and the role they play in the homeostatic processes in the healthy liver. In addition, we discuss the characteristic changes of EVs and their potential exacerbating or ameliorating effects in certain liver diseases, such as non-alcoholic fatty liver disease (NAFLD), alcoholic fatty liver disease (AFLD), drug induced liver injury (DILI), autoimmune hepatitis (AIH), hepatocarcinoma (HCC) and viral hepatitis.
Collapse
Affiliation(s)
- Viola Tamasi
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Krisztina Németh
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary
- ELKH-SE Translational Extracellular Vesicle Research Group, 1085 Budapest, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
8
|
Arakawa K, Inoue H, Ishigami A, Sato A, Takino Y, Tanaka M, Morimoto H, Takahashi N, Uehara M. Release of SMP30 in Extracellular Vesicles under Conditions of Ascorbic Acid Deficiency Is Involved with Acute Phase Response in ODS Rat. J Nutr Sci Vitaminol (Tokyo) 2023; 69:420-427. [PMID: 38171814 DOI: 10.3177/jnsv.69.420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Senescence marker protein-30 (SMP30) is a senescence marker molecule that exhibits lactonase activity in the ascorbic acid (AsA) biosynthesis pathway, except in primate mammals, including humans. Although numerous studies have shown that hepatic AsA deficiency causes acute-phase responses, details of the relationship between SMP30 expression and acute-phase responses in AsA-deficient conditions remain to be elucidated. Here, we investigated the effects of AsA deficiency on the relationship between SMP30 and acute liver injury in osteogenic disorder Shionogi (ODS) rats, which have a hereditary defect in AsA biosynthesis. Male-ODS rats (4 wk old) were pair-fed an AsA-free diet with distilled or 0.1% AsA-dissolved water for 14 d. Under AsA-deficient conditions, hepatic SMP30 protein level was decreased and liver injury markers, the serum aspartate aminotransferase/alanine transaminase ratio and cytokine-induced neutrophil chemoattractant-1 (CINC-1) concentration, were elevated. In contrast, SMP30 protein level in extracellular vesicles (EVs) was significantly increased in addition to the positive acute proteins haptoglobin and asialoglycoprotein receptor 1 (ASGPR1), hepatic-derived specific markers expression under AsA-deficient conditions. AsA deficiency also activated signal transducer and activator of transcription 3 (STAT3) which is linked to EVs release in the liver. These results suggest that the release of SMP30 in EVs by AsA deficiency is involved with acute-phase responses.
Collapse
Affiliation(s)
- Kohta Arakawa
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Hirofumi Inoue
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology
| | - Miori Tanaka
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Hiromu Morimoto
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Nobuyuki Takahashi
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Mariko Uehara
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture
| |
Collapse
|
9
|
Lu W, Tang H, Li S, Bai L, Chen Y. Extracellular vesicles as potential biomarkers and treatment options for liver failure: A systematic review up to March 2022. Front Immunol 2023; 14:1116518. [PMID: 36911706 PMCID: PMC9992400 DOI: 10.3389/fimmu.2023.1116518] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Extracellular vesicles (EVs) carrying functional cargoes are emerging as biomarkers and treatment strategies in multiple liver diseases. Nevertheless, the potential of EVs in liver failure remains indistinct. In this systematic review, we comprehensively analyzed the potential of EVs as biomarkers of liver failure and the therapeutic effects and possible mechanisms of EVs for liver failure. Methods We conducted a systematic review by comprehensively searching the following electronic databases: PubMed, Web of Science, Embase and Cochrane Central Register of Controlled Trials from inception to March 2022. The used text words (synonyms and word variations) and database-specific subject headings included "Extracellular Vesicles", "Exosomes", "Liver Failure", "Liver Injury", etc. Results A total of 1479 studies were identified. After removing 680 duplicate studies and 742 irrelevant studies, 57 studies were finally retained and analyzed. Fourteen studies revealed EVs with functional cargoes could be used to make the diagnosis of liver failure and provide clues for early warning and prognostic assessment of patients with liver failure. Forty-three studies confirmed the administration of EVs from different sources alleviated hepatic damage and improved survival through inhibiting inflammatory response, oxidative stress as well as apoptosis or promoting hepatocyte regeneration and autophagy. Conclusions EVs and their cargoes can be used not only as superior biomarkers of early warning, early diagnosis and prognostic assessments for liver failure, but also as potentially effective treatment options for liver failure. In the future, large-scale studies are urgently needed to verify the diagnostic, predictive and therapeutic value of EVs for liver failure.
Collapse
Affiliation(s)
- Wang Lu
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Huixin Tang
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Shanshan Li
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Li Bai
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| | - Yu Chen
- Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Beijing, China
| |
Collapse
|
10
|
Yeung CLS, Yam JWP. Therapy-induced modulation of extracellular vesicles in hepatocellular carcinoma. Semin Cancer Biol 2022; 86:1088-1101. [PMID: 35158067 DOI: 10.1016/j.semcancer.2022.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/27/2023]
Abstract
Despite rapid development of anti-tumorigenic treatments, the clinical outcome for hepatocellular carcinoma (HCC) is still far from satisfactory. With a deeper understanding about tumor microenvironment (TME), the critical role of extracellular vesicles (EVs) as intercellular liaison has come into spotlight. The dynamic functionality of these nanoparticles revealed cancer cells can employ both tumor and non-tumorous components for their own benefit, so as to mediate cell-to-cell communication and interchange of oncogenic biomolecules. Increasing studies on HCC-derived EVs have identified various irregulated biomolecules, that may serve as biomarkers or therapeutic targets. In this review, we first introduce the current knowledge about EVs and how they operate to maintain a healthy liver microenvironment. We then summarize some of the aberrant observations reported on HCC-derived EVs and how they contribute to HCC pathogenesis. Finally, we describe how current treatments for HCC alter behavior of EVs, which may shed light for potential prognostic markers and therapeutic strategies.
Collapse
Affiliation(s)
- Cherlie Lot Sum Yeung
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong.
| |
Collapse
|
11
|
Xie D, Qian B, Li X. Nucleic acids and proteins carried by exosomes from various sources: Potential role in liver diseases. Front Physiol 2022; 13:957036. [PMID: 36213232 PMCID: PMC9538374 DOI: 10.3389/fphys.2022.957036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022] Open
Abstract
Exosomes are extracellular membrane-encapsulated vesicles that are released into the extracellular space or biological fluids by many cell types through exocytosis. As a newly identified form of intercellular signal communication, exosomes mediate various pathological and physiological processes by exchanging various active substances between cells. The incidence and mortality of liver diseases is increasing worldwide. Therefore, we reviewed recent studies evaluating the role of exosomes from various sources in the diagnosis and treatment of liver diseases.
Collapse
Affiliation(s)
- Danna Xie
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Baolin Qian
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xun Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Department of General Surgery, the First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
- Center for Cancer Prevention and Treatment, School of Medicine, Lanzhou University, Lanzhou, China
- Gansu Provincial Institute of Hepatobiliary and Pancreatic Surgery, Lanzhou, China
- *Correspondence: Xun Li,
| |
Collapse
|
12
|
Abstract
Tissue engineering and regenerative medicine (TERM) may be defined as a translational discipline focused on the development of novel techniques, devices, and materials to replace or repair injured or diseased tissue and organs. The main approaches typically use cells, scaffolds, and signaling molecules, either alone or in combination, to promote repair and regeneration. Although cells are required to create new functional tissue, the source of cells, either from an exogenous allogeneic or autologous source or through the recruitment of endogenous (autologous) cells, is technically challenging and risks the host rejection of new tissue. Regardless of the cell source, these approaches also require appropriate instruction for proliferation, differentiation, and in vivo spatial organization to create new functional tissue. Such instruction is supplied through the microenvironment where cells reside, environments which largely consist of the extracellular matrix (ECM). The specific components of the ECM, and broadly the extracellular space, responsible for promoting tissue regeneration and repair, are not fully understood, however extracellular vesicles (EVs) found in body fluids and solid phases of ECM have emerged as key mediators of tissue regeneration and repair. Additionally, these EVs might serve as potential cell-free tools in TERM to promote tissue repair and regeneration with minimal risk for host rejection and adverse sequelae. The past two decades have shown a substantial interest in understanding the therapeutic role of EVs and their applications in the context of TERM. Therefore, the purpose of this review is to highlight the fundamental characteristics of EVs, the current pre-clinical and clinical applications of EVs in TERM, and the future of EV-based strategies in TERM.
Collapse
|
13
|
Wang J, Wang X, Zhang X, Shao T, Luo Y, Wang W, Han Y. Extracellular Vesicles and Hepatocellular Carcinoma: Opportunities and Challenges. Front Oncol 2022; 12:884369. [PMID: 35692794 PMCID: PMC9175035 DOI: 10.3389/fonc.2022.884369] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/25/2022] [Indexed: 12/05/2022] Open
Abstract
The incidence of hepatocellular carcinoma (HCC) is increasing worldwide. Extracellular vesicles (EVs) contain sufficient bioactive substances and are carriers of intercellular information exchange, as well as delivery vehicles for nucleic acids, proteins and drugs. Although EVs show great potential for the treatment of HCC and their role in HCC progression has been extensively studied, there are still many challenges such as time-consuming extraction, difficult storage, easy contamination, and low drug loading rate. We focus on the biogenesis, morphological characteristics, isolation and extraction of EVs and their significance in the progression of HCC, tumor invasion, immune escape and cancer therapy for a review. EVs may be effective biomarkers for molecular diagnosis of HCC and new targets for tumor-targeted therapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoya Wang
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xintong Zhang
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Tingting Shao
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yanmei Luo
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Wei Wang
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Academician (Expert) Workstation of Sichuan Province, Luzhou, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China.,School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
14
|
Weber B, Franz N, Marzi I, Henrich D, Leppik L. Extracellular vesicles as mediators and markers of acute organ injury: current concepts. Eur J Trauma Emerg Surg 2022; 48:1525-1544. [PMID: 33533957 PMCID: PMC7856451 DOI: 10.1007/s00068-021-01607-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Due to the continued high incidence and mortality rate worldwide, there is a need to develop new strategies for the quick, precise, and valuable recognition of presenting injury pattern in traumatized and poly-traumatized patients. Extracellular vesicles (EVs) have been shown to facilitate intercellular communication processes between cells in close proximity as well as distant cells in healthy and disease organisms. miRNAs and proteins transferred by EVs play biological roles in maintaining normal organ structure and function under physiological conditions. In pathological conditions, EVs change the miRNAs and protein cargo composition, mediating or suppressing the injury consequences. Therefore, incorporating EVs with their unique protein and miRNAs signature into the list of promising new biomarkers is a logical next step. In this review, we discuss the general characteristics and technical aspects of EVs isolation and characterization. We discuss results of recent in vitro, in vivo, and patients study describing the role of EVs in different inflammatory diseases and traumatic organ injuries. miRNAs and protein signature of EVs found in patients with acute organ injury are also debated.
Collapse
Affiliation(s)
- Birte Weber
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Niklas Franz
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Ingo Marzi
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Liudmila Leppik
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Fernandez-Checa JC, Bagnaninchi P, Ye H, Sancho-Bru P, Falcon-Perez JM, Royo F, Garcia-Ruiz C, Konu O, Miranda J, Lunov O, Dejneka A, Elfick A, McDonald A, Sullivan GJ, Aithal GP, Lucena MI, Andrade RJ, Fromenty B, Kranendonk M, Cubero FJ, Nelson LJ. Advanced preclinical models for evaluation of drug-induced liver injury - consensus statement by the European Drug-Induced Liver Injury Network [PRO-EURO-DILI-NET]. J Hepatol 2021; 75:935-959. [PMID: 34171436 DOI: 10.1016/j.jhep.2021.06.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of acute liver failure (ALF) and one of the leading indications for liver transplantation in Western societies. Given the wide use of both prescribed and over the counter drugs, DILI has become a major health issue for which there is a pressing need to find novel and effective therapies. Although significant progress has been made in understanding the molecular mechanisms underlying DILI, our incomplete knowledge of its pathogenesis and inability to predict DILI is largely due to both discordance between human and animal DILI in preclinical drug development and a lack of models that faithfully recapitulate complex pathophysiological features of human DILI. This is exemplified by the hepatotoxicity of acetaminophen (APAP) overdose, a major cause of ALF because of its extensive worldwide use as an analgesic. Despite intensive efforts utilising current animal and in vitro models, the mechanisms involved in the hepatotoxicity of APAP are still not fully understood. In this expert Consensus Statement, which is endorsed by the European Drug-Induced Liver Injury Network, we aim to facilitate and outline clinically impactful discoveries by detailing the requirements for more realistic human-based systems to assess hepatotoxicity and guide future drug safety testing. We present novel insights and discuss major players in APAP pathophysiology, and describe emerging in vitro and in vivo pre-clinical models, as well as advanced imaging and in silico technologies, which may improve prediction of clinical outcomes of DILI.
Collapse
Affiliation(s)
- Jose C Fernandez-Checa
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Consejo Superior Investigaciones Científicas (CSIC), Spain; Liver Unit, Hospital Clínic, Barcelona, Spain; Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, United States, CA 90033.
| | - Pierre Bagnaninchi
- Center for Regenerative Medicine, Institute for Regenerative and Repair, The University of Edinburgh, Edinburgh, UK, EH16 4UU; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Faraday Building, Colin Maclaurin Road, EH9 3 DW, Scotland, UK
| | - Hui Ye
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Pau Sancho-Bru
- Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Juan M Falcon-Perez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia, 48015, Spain
| | - Felix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, 48160, Spain
| | - Carmen Garcia-Ruiz
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Consejo Superior Investigaciones Científicas (CSIC), Spain; Liver Unit, Hospital Clínic, Barcelona, Spain; Instituto Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; USC Research Center for ALPD, Keck School of Medicine, Los Angeles, United States, CA 90033
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey; Interdisciplinary Neuroscience Program, Bilkent University, Ankara, Turkey; UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Joana Miranda
- Research Institute for iMedicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alistair Elfick
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh EH8 3DW, UK
| | - Alison McDonald
- Institute for Bioengineering, School of Engineering, The University of Edinburgh, Edinburgh EH8 3DW, UK
| | - Gareth J Sullivan
- University of Oslo and the Oslo University Hospital, Oslo, Norway; Hybrid Technology Hub-Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pediatric Research, Oslo University Hosptial, Oslo, Norway
| | - Guruprasad P Aithal
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospital NHS Trust and University of Nottingham, Nottingham, UK
| | - M Isabel Lucena
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, UICEC SCReN, Universidad de Málaga, Málaga, Spain
| | - Raul J Andrade
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Unidad de Gestión Clínica de Enfermedades Digestivas, Instituto de Investigación, Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Malaga, Spain
| | - Bernard Fromenty
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1241, F-35000 Rennes, France
| | - Michel Kranendonk
- Center for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, NOVA Medical School, Faculty of Medical Sciences, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Francisco Javier Cubero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, 28029, Spain; Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Leonard J Nelson
- Center for Regenerative Medicine, Institute for Regenerative and Repair, The University of Edinburgh, Edinburgh, UK, EH16 4UU; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Faraday Building, Colin Maclaurin Road, EH9 3 DW, Scotland, UK; Institute of Biological Chemistry, Biophysics and Bioengineering (IB3), School of Engineering and Physical Sciences (EPS), Heriot-Watt University, Edinburgh EH12 2AS, Scotland, UK.
| |
Collapse
|
16
|
Nakao Y, Amrollahi P, Parthasarathy G, Mauer AS, Sehrawat TS, Vanderboom P, Nair KS, Nakao K, Allen AM, Hu TY, Malhi H. Circulating extracellular vesicles are a biomarker for NAFLD resolution and response to weight loss surgery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 36:102430. [PMID: 34174416 DOI: 10.1016/j.nano.2021.102430] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/29/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022]
Abstract
There is increasing interest in the development of minimally invasive biomarkers for the diagnosis and prognosis of NAFLD via extracellular vesicles (EV). Plasma EVs were isolated by differential ultracentrifugation and quantified by nanoparticle tracking analysis from pre (n = 28) and post (n = 28) weight loss patients. In the pre weight loss group 22 had NAFLD. Nanoplasmon enhanced scattering (nPES) of gold nanoparticles conjugated to hepatocyte-specific antibodies was employed to identify hepatocyte-specific EVs. Complex lipid panel and targeted sphingolipids were performed. Logistic regression analysis was used to identify predictors of NAFLD. Plasma levels of EVs and hepatocyte-derived EVs are dynamic and decrease following NAFLD resolution due to weight loss surgery. Hepatocyte-derived EVs correlate with steatosis in NAFLD patients and steatosis and inflammation in NASH patients. Plasma levels of small EVs correlate with EV sphingolipids in patients with NASH. Hepatocyte-derived EVs measured by the nPES assay could serve as a point-of-care test for NAFLD.
Collapse
Affiliation(s)
- Yasuhiko Nakao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN; Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Pouya Amrollahi
- Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | | | - Amy S Mauer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | | | | | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Alina M Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Tony Y Hu
- Virginia G. Piper Biodesign Center for Personalized Diagnostics, The Biodesign Institute, Arizona State University, Tempe, AZ, USA; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
17
|
Kovač Peić A, Šrajer Gajdošik M, Brilliant K, Callanan H, Hixson DC, Begić M, Josić D. Changes in the proteome of extracellular vesicles shed by rat liver after subtoxic exposure to acetaminophen. Electrophoresis 2021; 42:1388-1398. [PMID: 33837589 DOI: 10.1002/elps.202100020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 01/16/2023]
Abstract
To identify changes in extracellular vesicles (EVs) secreted by the liver following drug-induced liver injury (DILI), rats were treated with a subtoxic dose (500 mg/kg) of the analgesic drug, acetaminophen (APAP). EVs were collected by liver perfusion of sham and APAP-treated rats. Changes in EVs morphology were examined by transmission electron microscopic analysis of negatively stained vesicles. Results from morphometric analysis of EVs revealed striking differences in their size and distribution. Proteome composition of EVs collected by liver perfusion was determined by mass spectrometry using methods of sample preparation that enabled better detection of both highly hydrophobic proteins and proteins with complex post-translational modifications. The collection of EVs after liver perfusion is an approach that enables the isolation of EVs shed not only by isolated hepatocytes, but also by the entire complement of hepatic cells. EVs derived after DILI had a lower content of alpha-1-macroglobulin, ferritin, and members of cytochrome 450 family. Fibronectin, aminopeptidase N, metalloreductase STEAP4, integrin beta, and members of the annexin family were detected only in APAP-treated samples of EVs. These results show that the present approach can provide valuable insights into the response of the liver following drug-induced liver injury.
Collapse
Affiliation(s)
| | | | - Kate Brilliant
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI, USA
| | - Helen Callanan
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI, USA
| | - Douglas C Hixson
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI, USA.,Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Marija Begić
- Faculty of Medicine, Juraj Dobrila University of Pula, Pula, Croatia
| | - Djuro Josić
- Proteomics Core, COBRE CCRD, Rhode Island Hospital, Providence, RI, USA.,Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
18
|
Gonzalez E, Azkargorta M, Garcia-Vallicrosa C, Prieto-Elordui J, Elortza F, Blanco-Sampascual S, Falcon-Perez JM. Could protein content of Urinary Extracellular Vesicles be useful to detect Cirrhosis in Alcoholic Liver Disease? Int J Biol Sci 2021; 17:1864-1877. [PMID: 34131392 PMCID: PMC8193259 DOI: 10.7150/ijbs.59725] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/10/2021] [Indexed: 12/13/2022] Open
Abstract
Alcohol abuse has a high impact on the mortality and morbidity related to a great number of diseases and is responsible for the development of alcoholic liver disease (ALD). It remains challenging to detect and evaluate its severity, which is crucial for prognosis. In this work, we studied if urinary EVs (uEVs) could serve in diagnose and evaluate cirrhosis in ALD. To this purpose, uEVs characterization by cryo-electron microscopy (Cryo-EM), Nanoparticle Tracking Analysis (NTA) and Western blotting (WB) was performed in a cohort of 21 controls and 21 cirrhotic patients. Then, proteomics of uEVs was carried out in a second cohort of 6 controls and 8 patients in order to identify new putative biomarkers for cirrhosis in ALD. Interestingly, uEVs concentration, size and protein composition were altered in cirrhotic patients. From a total of 1304 proteins identified in uEVs, 90 of them were found to be altered in cirrhotic patients. The results suggest that uEVs could be considered as a tool and a supplier of new biomarkers for cirrhosis in ALD, whose application would be especially relevant in chronic patients. Yet, further research is necessary to obtain more relevant result in clinical terms.
Collapse
Affiliation(s)
- Esperanza Gonzalez
- Exosomes Laboratory. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Mikel Azkargorta
- Proteomics Platform. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Clara Garcia-Vallicrosa
- Exosomes Laboratory. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Felix Elortza
- Proteomics Platform. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | | | - Juan Manuel Falcon-Perez
- Exosomes Laboratory. Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Madrid, Spain
- IKERBASQUE Basque Foundation for Science Bilbao Spain
| |
Collapse
|
19
|
Zivko C, Fuhrmann G, Luciani P. Liver-derived extracellular vesicles: A cell by cell overview to isolation and characterization practices. Biochim Biophys Acta Gen Subj 2021; 1865:129559. [DOI: 10.1016/j.bbagen.2020.129559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/16/2020] [Accepted: 02/11/2020] [Indexed: 02/08/2023]
|
20
|
Kim HJ, Kim OH, Hong HE, Lee SC, Kim SJ. Harnessing adipose‑derived stem cells to release specialized secretome for the treatment of hepatitis B. Int J Mol Med 2021; 47:15. [PMID: 33448314 PMCID: PMC7834954 DOI: 10.3892/ijmm.2021.4848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 12/09/2020] [Indexed: 11/09/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have the function of repairing damaged tissue, which is known to be mediated by the secretome, the collection of secretory materials shed from MSCs. Adjusting the culture conditions of MSCs can lead to a significant difference in the composition of the secretome. It was hypothesized that pre‑sensitization of MSCs with specific disease‑causing agents could harness MSCs to release the therapeutic materials specialized for the disease. To validate this hypothesis, the present study aimed to generate a 'disease‑specific secretome' for hepatitis caused by hepatitis B virus using hepatitis BX antigen (HBx) as a disease‑causing material. Secretary materials (HBx‑IS) were collected following the stimulation of adipose‑derived stem cells (ASCs) with 100‑fold diluted culture media of AML12 hepatocytes that had been transfected with pcDNA‑HBx for 24 h. An animal model of hepatitis B was generated by injecting HBx into mice, and the mice were subsequently intravenously administered a control secretome (CS) or HBx‑IS. Compared with the CS injection, the HBx‑IS injection significantly reduced the serum levels of interleukin‑6 and tumor necrosis factor‑α (pro‑inflammatory cytokines). Western blot analysis and immunohistochemistry of the liver specimens revealed that the HBx‑IS injection led to a higher expression of liver regeneration‑related markers, including hepatocyte growth factor and proliferating cell nuclear antigen, a lower expression of pro‑apoptotic markers, such as cleaved caspase 3 and Bim in mouse livers, and a lower expression of pro‑inflammatory markers (F4/80 and CD68) compared to the CS injection. HBx‑IS exhibited higher liver regenerative, anti‑inflammatory and anti‑apoptotic properties, particularly in the mouse model of hepatitis B compared to CS. This suggests that the secretome obtained by stimulating ASCs with disease‑causing agents may have a more prominent therapeutic effect on the specific disease than the naïve secretome.
Collapse
Affiliation(s)
- Hee Ju Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591
| | - Ok-Hee Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea
| | - Ha-Eun Hong
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea
| | - Sang Chul Lee
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon 34943, Republic of Korea
| | - Say-June Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea
| |
Collapse
|
21
|
Zhao L, Wang Y, Zhang Y. The potential diagnostic and therapeutic applications of exosomes in drug-induced liver injury. Toxicol Lett 2020; 337:68-77. [PMID: 33259895 DOI: 10.1016/j.toxlet.2020.11.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) has gradually become a global public medical problem, which can be caused by more than 1000 currently available drugs. Unfortunately, the diagnosis and treatment of DILI are limited and imperfect. Exosomes can be secreted by a variety of cells and tissues in the body, rich in cell-type specific proteins, nucleic acids and lipids, which has been widely studied as an important intercellular communication vehicle in liver diseases. Emerging data suggest that circulating exosomes and their cargos can be used as minimally-invasive sources of potential molecular biomarkers for the early detection, monitoring and evaluation of DILI. Exosomes in the urine were also found to contain proteins or RNAs that were indicative of DILI. In addition, exosomes derived from mesenchymal stem cell or hepatocyte are considered potential therapeutic agents to promote liver regenerative responses, modulate inflammatory response and deduce hepatocytes apoptosis. Based on the current findings, we suggest the potential applications of exosomes as biomarkers and therapeutics for DILI.
Collapse
Affiliation(s)
- Lanlan Zhao
- Department of Gerontology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuezhi Wang
- Department of Gerontology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of Gerontology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
van Meteren N, Lagadic-Gossmann D, Podechard N, Gobart D, Gallais I, Chevanne M, Collin A, Burel A, Dupont A, Rault L, Chevance S, Gauffre F, Le Ferrec E, Sergent O. Extracellular vesicles released by polycyclic aromatic hydrocarbons-treated hepatocytes trigger oxidative stress in recipient hepatocytes by delivering iron. Free Radic Biol Med 2020; 160:246-262. [PMID: 32791186 DOI: 10.1016/j.freeradbiomed.2020.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/14/2020] [Accepted: 08/01/2020] [Indexed: 12/11/2022]
Abstract
A growing body of evidences indicate the major role of extracellular vesicles (EVs) as players of cell communication in the pathogenesis of liver diseases. EVs are membrane-enclosed vesicles released by cells into the extracellular environment. Oxidative stress is also a key component of liver disease pathogenesis, but no role for hepatocyte-derived EVs has yet been described in the development of this process. Recently, some polycyclic aromatic hydrocarbons (PAHs), widespread environmental contaminants, were demonstrated to induce EV release from hepatocytes. They are also well-known to trigger oxidative stress leading to cell death. Therefore, the aim of this work was to investigate the involvement of EVs derived from PAHs-treated hepatocytes (PAH-EVs) in possible oxidative damages of healthy recipient hepatocytes, using both WIF-B9 and primary rat hepatocytes. We first showed that the release of EVs from PAHs -treated hepatocytes depended on oxidative stress. PAH-EVs were enriched in proteins related to oxidative stress such as NADPH oxidase and ferritin. They were also demonstrated to contain more iron. PAH-EVs could then induce oxidative stress in recipient hepatocytes, thereby leading to apoptosis. Mitochondria and lysosomes of recipient hepatocytes exhibited significant structural alterations. All those damages were dependent on internalization of EVs that reached lysosomes with their cargoes. Lysosomes thus appeared as critical organelles for EVs to induce apoptosis. In addition, pro-oxidant components of PAH-EVs, e.g. NADPH oxidase and iron, were revealed to be necessary for this cell death.
Collapse
Affiliation(s)
- Nettie van Meteren
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Normand Podechard
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Dimitri Gobart
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Isabelle Gallais
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Martine Chevanne
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Aurore Collin
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Agnès Burel
- Univ Rennes, Biosit, UMS 3480, US_S 018, F-35000, Rennes, France
| | - Aurélien Dupont
- Univ Rennes, Biosit, UMS 3480, US_S 018, F-35000, Rennes, France
| | | | - Soizic Chevance
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Fabienne Gauffre
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000, Rennes, France
| | - Eric Le Ferrec
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Odile Sergent
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| |
Collapse
|
23
|
Umbaugh DS, Jaeschke H. Extracellular vesicles: Roles and applications in drug-induced liver injury. Adv Clin Chem 2020; 102:63-125. [PMID: 34044913 DOI: 10.1016/bs.acc.2020.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EV) are defined as nanosized particles, with a lipid bilayer, that are unable to replicate. There has been an exponential increase of research investigating these particles in a wide array of diseases and deleterious states (inflammation, oxidative stress, drug-induced liver injury) in large part due to increasing recognition of the functional capacity of EVs. Cells can package lipids, proteins, miRNAs, DNA, and RNA into EVs and send these discrete packages of molecular information to distant, recipient cells to alter the physiological state of that cell. EVs are innately heterogeneous as a result of the diverse molecular pathways that are used to generate them. However, this innate heterogeneity of EVs is amplified due to the diversity in isolation techniques and lack of standardized nomenclature in the literature making it unclear if one scientist's "exosome" is another scientist's "microvesicle." One goal of this chapter is to provide the contextual understanding of EV origin so one can discern between divergent nomenclature. Further, the chapter will explore the potential protective and harmful roles that EVs play in DILI, and the potential of EVs and their cargo as a biomarker. The use of EVs as a therapeutic as well as a vector for therapeutic delivery will be discussed.
Collapse
Affiliation(s)
- David S Umbaugh
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
24
|
Azparren-Angulo M, Royo F, Gonzalez E, Liebana M, Brotons B, Berganza J, Goñi-de-Cerio F, Manicardi N, Abad-Jordà L, Gracia-Sancho J, Falcon-Perez JM. Extracellular vesicles in hepatology: Physiological role, involvement in pathogenesis, and therapeutic opportunities. Pharmacol Ther 2020; 218:107683. [PMID: 32961265 DOI: 10.1016/j.pharmthera.2020.107683] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Since the first descriptions of hepatocyte-released exosome-like vesicles in 2008, the number of publications describing Extracellular Vesicles (EVs) released by liver cells in the context of hepatic physiology and pathology has grown exponentially. This growing interest highlights both the importance that cell-to-cell communication has in the organization of multicellular organisms from a physiological point of view, as well as the opportunity that these circulating organelles offer in diagnostics and therapeutics. In the present review, we summarize systematically and comprehensively the myriad of works that appeared in the last decade and lighted the discussion about the best opportunities for using EVs in liver disease therapeutics.
Collapse
Affiliation(s)
- Maria Azparren-Angulo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Felix Royo
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain; Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Esperanza Gonzalez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Marc Liebana
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Bruno Brotons
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain
| | - Jesús Berganza
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Parque Tecnológico, Edificio 202, 48170 Zamudio, Bizkaia, Spain
| | - Felipe Goñi-de-Cerio
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Parque Tecnológico, Edificio 202, 48170 Zamudio, Bizkaia, Spain
| | - Nicoló Manicardi
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Laia Abad-Jordà
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS, CIBEREHD, Barcelona, Spain; Hepatology, Department of Biomedical Research, Inselspital & University of Bern, Switzerland
| | - Juan M Falcon-Perez
- Exosomes Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia 48160, Spain; Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid 28029, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Bizkaia 48015, Spain.
| |
Collapse
|
25
|
Soekmadji C, Li B, Huang Y, Wang H, An T, Liu C, Pan W, Chen J, Cheung L, Falcon-Perez JM, Gho YS, Holthofer HB, Le MTN, Marcilla A, O'Driscoll L, Shekari F, Shen TL, Torrecilhas AC, Yan X, Yang F, Yin H, Xiao Y, Zhao Z, Zou X, Wang Q, Zheng L. The future of Extracellular Vesicles as Theranostics - an ISEV meeting report. J Extracell Vesicles 2020; 9:1809766. [PMID: 33144926 PMCID: PMC7580849 DOI: 10.1080/20013078.2020.1809766] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The utilization of extracellular vesicles (EVs) in clinical theranostics has rapidly advanced in the past decade. In November 2018, the International Society for Extracellular Vesicles (ISEV) held a workshop on “EVs in Clinical Theranostic”. Here, we report the conclusions of roundtable discussions on the current advancement in the analysis technologies and we provide some guidelines to researchers in the field to consider the use of EVs in clinical application. The main challenges and the requirements for EV separation and characterization strategies, quality control and clinical investigation were discussed to promote the application of EVs in future clinical studies.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Bo Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haifang Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Taixue An
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunchen Liu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weilun Pan
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Chen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lesley Cheung
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Juan Manuel Falcon-Perez
- Exosomes Laboratory and Metabolomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain.,Centro De Investigación Biomédica En Red De Enfermedades Hepáticas Y Digestivas (Ciberehd), Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Yong Song Gho
- Laboratory of Intercellular Communication, Department of Life Science, POSTECH, South Korea
| | - Harry B Holthofer
- Medical Department, University Medical Center Hamburg-Eppendorf, Germany
| | - Minh T N Le
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Antonio Marcilla
- Àrea De Parasitologia, Departament De Farmàcia I Tecnologia Farmacèutica I Parasitologia, Universitat De València, Burjassot, Valencia, Spain.,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute La Fe-Universitat De Valencia, Valencia, Spain
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute & Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Trinity St. James's Cancer Institute (TSJCI), Trinity College Dublin, Dublin, Ireland
| | - Faezeh Shekari
- Department of Stem Cells and Developmental BiologyCell Science, Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tang Long Shen
- Department of Plant Pathology and Microbiology & Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | | | - Xiaomei Yan
- Department of Chemical Biology, the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian, China
| | - Fuquan Yang
- Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yu Xiao
- Laboratory of Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zezhou Zhao
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xue Zou
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Qian Wang
- Laboratory of Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Li Y, Zhu X, Wang G, Tong H, Su L, Li X. Proteomic analysis of extracellular vesicles released from heat-stroked hepatocytes reveals promotion of programmed cell death pathway. Biomed Pharmacother 2020; 129:110489. [PMID: 32768969 DOI: 10.1016/j.biopha.2020.110489] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Liver injury is a common complication of severe heat stroke (HS). Extracellular vesicles (EVs) are part of a novel pathway mediating intercellular communication. Whether EVs are involved in the pathogenesis underlying HS-induced liver injury remains unknown. Here, we explored the role of hepatocyte EVs in HS-induced liver injury and their protein regulation patterns after HS induction. Isobaric tags for relative and absolute quantification technology (iTRAQ) and liquid chromatography-tandem mass spectrometry analysis identified changes in the proteomic profiles of hepatocyte-derived heat-stroked EVs, and we identified 53 up-regulated proteins. Bioinformatics analysis verified that the regulation of programmed cell death was the most significant altered pathway. To clarify the effect of HS hepatocyte-derived EVs in inducing hepatocyte-programmed death and injury, they were added to recipient hepatocytes and injected into mice. This treatment significantly induced the synthesis of apoptosis (caspase-3/8) and necroptosis-associated proteins [receptor-interacting protein 1 (RIP1), RIP3, and mixed lineage kinase domain-like protein]; moreover, it increased the numbers of apoptotic and necroptotic cells in hepatocytes and liver tissues and increased the levels of biochemical liver injury markers (alanine aminotransferase, aspartate aminotransferase, and lactate dehydrogenase). Our study is the first comprehensive analysis of the hepatocyte-derived heat-stroked EV proteome confirming the induction of liver injury by Evs. We provide a novel explanation for the pathological mechanism underlying HS-induced liver injury.
Collapse
Affiliation(s)
- Yue Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Intensive Care Unit, Guangzhou General Hospital of Guangzhou Military Command, Southern Medical University, Guangzhou, China.
| | - Xintao Zhu
- Department of Intensive Care Unit, Guangzhou General Hospital of Guangzhou Military Command, Southern Medical University, Guangzhou, China.
| | - Guozhen Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Huasheng Tong
- Department of Intensive Care Unit, Guangzhou General Hospital of Guangzhou Military Command, Southern Medical University, Guangzhou, China.
| | - Lei Su
- Department of Intensive Care Unit, Guangzhou General Hospital of Guangzhou Military Command, Southern Medical University, Guangzhou, China.
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
27
|
Povero D, Yamashita H, Ren W, Subramanian MG, Myers RP, Eguchi A, Simonetto DA, Goodman ZD, Harrison SA, Sanyal AJ, Bosch J, Feldstein AE. Characterization and Proteome of Circulating Extracellular Vesicles as Potential Biomarkers for NASH. Hepatol Commun 2020; 4:1263-1278. [PMID: 32923831 PMCID: PMC7471415 DOI: 10.1002/hep4.1556] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/17/2020] [Accepted: 05/11/2020] [Indexed: 12/25/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is currently one of most common forms of chronic liver disease globally. NAFLD represents a wide spectrum of liver involvement from nonprogressive isolated steatosis to nonalcoholic steatohepatitis (NASH), characterized by liver necroinflammation and fibrosis and currently one of the top causes of end‐stage liver disease and hepatocellular carcinoma. At present, there is a lack of effective treatments, and a central barrier to the development of therapies is the requirement for an invasive liver biopsy for diagnosis of NASH. Discovery of reliable, noninvasive biomarkers are urgently needed. In this study, we tested whether circulating extracellular vesicles (EVs), cell‐derived small membrane‐surrounded structures with a rich cargo of bioactive molecules, may serve as reliable noninvasive “liquid biopsies” for NASH diagnosis and assessment of disease severity. Total circulating EVs and hepatocyte‐derived EVs were isolated by differential centrifugation and size‐exclusion chromatography from serum samples of healthy individuals, patients with precirrhotic NASH, and patients with cirrhotic NASH. EVs were further characterized by flow cytometry, electron microscopy, western blotting, and dynamic light scattering assays before performing a proteomics analysis. Our findings suggest that levels of total and hepatocyte‐derived EVs correlate with NASH clinical characteristics and disease severity. Additionally, using proteomics data, we developed understandable, powerful, and unique EV‐based proteomic signatures for potential diagnosis of advanced NASH. Conclusion: Our study shows that the quantity and protein constituents of circulating EVs provide strong evidence for EV protein–based liquid biopsies for NAFLD/NASH diagnosis.
Collapse
Affiliation(s)
- Davide Povero
- Department of Pediatrics University of California San Diego La Jolla CA
| | | | - Wenhua Ren
- Genomics and Microarray Core University of Colorado Denver Aurora CO
| | | | | | - Akiko Eguchi
- Department of Pediatrics University of California San Diego La Jolla CA
| | | | | | | | | | - Jaime Bosch
- Inselspital Bern University Bern Switzerland.,Ciberehd-Idibaps University of Barcelona Barcelona Spain
| | - Ariel E Feldstein
- Department of Pediatrics University of California San Diego La Jolla CA
| |
Collapse
|
28
|
Shabangu CS, Huang JF, Hsiao HH, Yu ML, Chuang WL, Wang SC. Liquid Biopsy for the Diagnosis of Viral Hepatitis, Fatty Liver Steatosis, and Alcoholic Liver Diseases. Int J Mol Sci 2020; 21:3732. [PMID: 32466319 PMCID: PMC7279404 DOI: 10.3390/ijms21103732] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
During the progression from hepatitis to fibrosis, cirrhosis, and liver failure, the accumulation of stressed/damaged hepatocyte elements associated with liver inflammation is critical. The causes of hepatocyte injuries include viral hepatitis infections, alcoholic hepatitis, and non-alcoholic fatty liver disease. Hepatocyte-derived extracellular vesicles (Hep-EVs) released from stressed/damaged hepatocytes are partly responsible for liver disease progression and liver damage because they activate non-parenchymal cells and infiltrate inflammatory cells within the liver, which are in turn are an important source of EVs. This cell-to-cell signaling is prevalent during inflammation in many liver diseases. Accordingly, special emphasis should be placed on liquid biopsy methods for the long-term monitoring of chronic liver diseases. In the present review, we have highlighted various aspects of current liquid biopsy research into chronic liver diseases. We have also reviewed recent progress on liquid biopsies that focus on cell-free DNA (cfDNA), long non-coding RNA (lncRNA), and the proteins in EVs as potential diagnostic tools and novel therapeutic targets in patients with viral hepatitis, fatty liver steatosis, and alcoholic liver diseases.
Collapse
Affiliation(s)
- Ciniso Sylvester Shabangu
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-F.H.); (M.-L.Y.)
| | - Jee-Fu Huang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-F.H.); (M.-L.Y.)
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Faculty of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hui-Hua Hsiao
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ming-Lung Yu
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-F.H.); (M.-L.Y.)
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Faculty of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Hepatitis Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wan-Long Chuang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Faculty of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (J.-F.H.); (M.-L.Y.)
- Center for Liquid Biopsy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| |
Collapse
|
29
|
Kim OH, Hong HE, Seo H, Kwak BJ, Choi HJ, Kim KH, Ahn J, Lee SC, Kim SJ. Generation of induced secretome from adipose-derived stem cells specialized for disease-specific treatment: An experimental mouse model. World J Stem Cells 2020; 12:70-86. [PMID: 32110276 PMCID: PMC7031761 DOI: 10.4252/wjsc.v12.i1.70] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/16/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recently, the exclusive use of mesenchymal stem cell (MSC)-secreted molecules, named as the secretome, have been evaluated for overcoming the limitations of cell-based therapy while maintaining its advantages.
AIM To improve cell-free therapy by adding disease-specificity through stimulation of MSCs using disease-causing materials.
METHODS We collected the secretory materials (named as inducers) released from AML12 hepatocytes that had been pretreated with thioacetamide (TAA) and generated the TAA-induced secretome (TAA-isecretome) after stimulating adipose-derived stem cells with the inducers. The TAA-isecretome was intravenously administered to mice with TAA-induced hepatic failure and those with partial hepatectomy.
RESULTS TAA-isecretome infusion showed higher therapeutic potential in terms of (1) restoring disorganized hepatic tissue to normal tissue; (2) inhibiting proinflammatory cytokines (interleukin-6 and tumor necrosis factor-α); and (3) reducing abnormally elevated liver enzymes (aspartate aminotransferase and alanine aminotransferase) compared to the naïve secretome infusion in mice with TAA-induced hepatic failure. However, the TAA-isecretome showed inferior therapeutic potential for restoring hepatic function in partially hepatectomized mice. Proteomic analysis of TAA-isecretome identified that antioxidant processes were the most predominant enriched biological networks of the proteins exclusively identified in the TAA-isecretome. In addition, peroxiredoxin-1, a potent antioxidant protein, was found to be one of representative components of TAA-isecretome and played a central role in the protection of TAA-induced hepatic injury.
CONCLUSION Appropriate stimulation of adipose-derived stem cells with TAA led to the production of a secretome enriched with proteins, especially peroxiredoxin-1, with higher antioxidant activity. Our results suggest that appropriate stimulation of MSCs with pathogenic agents can lead to the production of a secretome specialized for protecting against the pathogen. This approach is expected to open a new way of developing various specific therapeutics based on the high plasticity and responsiveness of MSCs.
Collapse
Affiliation(s)
- Ok-Hee Kim
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| | - Ha-Eun Hong
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| | - Haeyeon Seo
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| | - Bong Jun Kwak
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| | - Ho Joong Choi
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| | - Kee-Hwan Kim
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, the Catholic University of Korea, Seoul 11765, South Korea
| | - Joseph Ahn
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| | - Sang Chul Lee
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
- Department of Surgery, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 34943, South Korea
| | - Say-June Kim
- Catholic Central Laboratory of Surgery, Institute of Biomedical Industry, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
- Department of Surgery, Division of Hepato-biliary Pancreatic Surgery, Seoul St. Mary’s Hospital, College of Medicine, the Catholic University of Korea, Seoul 06591, South Korea
| |
Collapse
|
30
|
Circulating Extracellular Vesicles Containing Xenobiotic Metabolizing CYP Enzymes and Their Potential Roles in Extrahepatic Cells Via Cell-Cell Interactions. Int J Mol Sci 2019; 20:ijms20246178. [PMID: 31817878 PMCID: PMC6940889 DOI: 10.3390/ijms20246178] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
The cytochrome P450 (CYP) family of enzymes is known to metabolize the majority of xenobiotics. Hepatocytes, powerhouses of CYP enzymes, are where most drugs are metabolized into non-toxic metabolites. Additional tissues/cells such as gut, kidneys, lungs, blood, and brain cells express selective CYP enzymes. Extrahepatic CYP enzymes, especially in kidneys, also metabolize drugs into excretable forms. However, extrahepatic cells express a much lower level of CYPs than hepatocytes. It is possible that the liver secretes CYP enzymes, which circulate via plasma and are eventually delivered to extrahepatic cells (e.g., brain cells). CYP circulation likely occurs via extracellular vesicles (EVs), which carry important biomolecules for delivery to distant cells. Recent studies have revealed an abundance of several CYPs in plasma EVs and other cell-derived EVs, and have demonstrated the role of CYP-containing EVs in xenobiotic-induced toxicity via cell–cell interactions. Thus, it is important to study the mechanism for packaging CYP into EVs, their circulation via plasma, and their role in extrahepatic cells. Future studies could help to find novel EV biomarkers and help to utilize EVs in novel interventions via CYP-containing EV drug delivery. This review mainly covers the abundance of CYPs in plasma EVs and EVs derived from CYP-expressing cells, as well as the potential role of EV CYPs in cell–cell communication and their application with respect to novel biomarkers and therapeutic interventions.
Collapse
|
31
|
Balaphas A, Meyer J, Sadoul R, Morel P, Gonelle-Gispert C, Bühler LH. Extracellular vesicles: Future diagnostic and therapeutic tools for liver disease and regeneration. Liver Int 2019; 39:1801-1817. [PMID: 31286675 DOI: 10.1111/liv.14189] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/06/2019] [Accepted: 07/01/2019] [Indexed: 02/13/2023]
Abstract
Extracellular vesicles are membrane fragments that can be produced by all cell types. Interactions between extracellular vesicles and various liver cells constitute an emerging field in hepatology and recent evidences have established a role for extracellular vesicles in various liver diseases and physiological processes. Extracellular vesicles originating from liver cells are implicated in intercellular communication and fluctuations of specific circulating extracellular vesicles could constitute new diagnostic tools. In contrast, extracellular vesicles derived from progenitor cells interact with hepatocytes or non-parenchymal cells, thereby protecting the liver from various injuries and promoting liver regeneration. Our review focuses on recent developments investigating the role of various types of extracellular vesicles in acute and chronic liver diseases as well as their potential use as biomarkers and therapeutic tools.
Collapse
Affiliation(s)
- Alexandre Balaphas
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland.,Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland.,Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Jeremy Meyer
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland.,Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland.,Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Rémy Sadoul
- Université Grenoble Alpes, Institut des Neurosciences, Grenoble, France
| | - Philippe Morel
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland.,Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland.,Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland.,Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Leo Hans Bühler
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland.,Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland.,Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
32
|
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]
|
33
|
Changes of Circulating Extracellular Vesicles from the Liver after Roux-en-Y Bariatric Surgery. Int J Mol Sci 2019; 20:ijms20092153. [PMID: 31052333 PMCID: PMC6539504 DOI: 10.3390/ijms20092153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 12/19/2022] Open
Abstract
Circulating extracellular vesicles are small particles enclosed by a phospholipid bilayer. Vesicles deriving directly from the cellular membrane by an active budding process retain cell origin specific proteins and RNA. These vesicles carry pathophysiological information from their parental cell and hold the potential to allow analysis of organs without the need for a biopsy. We included in our study 27 patients undergoing bariatric surgery. Hepatic extracellular vesicles were determined by flow cytometry. mRNA specific for hepatic cellular origin was determined in the extracellular vesicle fraction using qPCR. Surgery led to a massive reduction of weight and overall hepatic stress as determined by alanine transaminase (ALT), aspartate transaminase (AST) and γ-glutamyltransferase (GGT). Total extracellular vesicle numbers were reduced after bariatric surgery. Liver specific vesicles identified by HepPar1 or asialoglycoprotein receptor (ASGPR) were significantly reduced after bariatric surgery in both AnnexinV+ and AnnexinV− subgroups. When analyzing circulating liver-specific mRNAs, we found reduced levels of these mRNAs after surgery even though total circulating RNA remained unchanged. We conclude that circulating hepatic extracellular vesicles are detectable in samples from patients undergoing gastric bypass surgery. These vesicles are reduced after a reduction of hepatic stress also observed with classic liver enzyme measurements. We conclude that ASGPR or HepPar positive vesicles hold the potential to serve as liver specific vesicle markers.
Collapse
|
34
|
Royo F, Gil-Carton D, Gonzalez E, Mleczko J, Palomo L, Perez-Cormenzana M, Mayo R, Alonso C, Falcon-Perez JM. Differences in the metabolite composition and mechanical properties of extracellular vesicles secreted by hepatic cellular models. J Extracell Vesicles 2019; 8:1575678. [PMID: 30788084 PMCID: PMC6374943 DOI: 10.1080/20013078.2019.1575678] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/19/2018] [Accepted: 01/18/2019] [Indexed: 01/09/2023] Open
Abstract
Liver constitutes the major metabolic factory in the organism and is involved in the synthesis, secretion and clearance of many blood-circulating molecules. Previously, we have characterised the protein and RNA cargo of extracellular vesicles (EVs) secreted by two hepatic cellular models, a mouse hepatocyte progenitor cell line (MLP29) and primary rat hepatocytes (RHs). Here, we report the metabolome profile of these vesicles by performing a targeted UHPLC-MS metabolomics analysis of these two cellular models and their respective secreted EVs. Visual inspection of the data through principal component analysis allows clear separation of the metabolic profile of cells and EVs, and also of both cellular models. Correlation matrix supported that lipid composition of EVs is mainly determined by parent cell composition. EVs derived from MLP29 and RHs showed a negative correlation in their percentage composition of ceramides, glycerophospholipids, sphingomyelins and triglycerides. Metabolites enriched in EVs were also different depending on the cellular model. EVs secreted by MLP29 were enriched in different species of sphingomyelins and ceramides underrepresented in EVs secreted by RHs. Remarkably, triglycerides constitute an important percentage of the composition of EVs derived from RHs. We further investigate if the differences in lipid composition were also accompanied by differences in mechanical behaviour, by using atomic force microscopy complemented with nanoindentation-based methodology. To compare the stiffness and brittleness of EVs derived from MLP29 cell line and RH primary cells, FZ curves were performed in the centre of single vesicles and the differences found in their force-vs.-indentation curves suggest that RHs EVs are softer (less stiff) and less resistance to mechanical failure than MLP29 EVs. Therefore, we can conclude that EVs from different origin carry a characteristic lipid composition related to their parental cell composition, and exhibit different mechanical properties. Abbreviations: For the identification of the different species of lipids, the following abbreviations has been employed: Cer, ceramide; ChoE, Cholesteryl Ester; CMH, monohexosylceramide; DAG, diglycerid; LPC, lysophosphatidylcholin; LPI, lysophosphatidyinositol; PC, phosphocoline; PE, phoethanolamine; PI, phosphoinositol; SM, sphingomyelin; TAG, triglycerid
Collapse
Affiliation(s)
- Felix Royo
- Exosomes Laboratory, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Spain
| | - David Gil-Carton
- Electron Microscopy Technology Platform, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
| | | | - Justyna Mleczko
- Exosomes Laboratory, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
| | - Laura Palomo
- Exosomes Laboratory, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain
| | | | - Rebeca Mayo
- OWL Metabolomics, Bizkaia Technology Park, Derio, Spain
| | | | - Juan M Falcon-Perez
- Exosomes Laboratory, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Spain.,Metabolomics platform, CIC bioGUNE, Bizkaia Technology Park, Derio, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
35
|
Palomo L, Mleczko JE, Azkargorta M, Conde-Vancells J, González E, Elortza F, Royo F, Falcon-Perez JM. Abundance of Cytochromes in Hepatic Extracellular Vesicles Is Altered by Drugs Related With Drug-Induced Liver Injury. Hepatol Commun 2018; 2:1064-1079. [PMID: 30202821 PMCID: PMC6128234 DOI: 10.1002/hep4.1210] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022] Open
Abstract
Drug‐induced liver injury (DILI) is a serious worldwide health problem that accounts for more than 50% of acute liver failure. There is a great interest in clinical diagnosis and pharmaceutical industry to elucidate underlying molecular mechanisms and find noninvasive biomarkers for this pathology. Cell‐secreted extracellular vesicles (EVs) have provided a new biological source to identify low disease invasive markers. Despite the intense research developed on these vesicles, there is currently a gap on their patho‐physiological effects. Here, we study EVs secreted by primary rat hepatocytes challenged with galactatosamine (GalN), acetaminophen, or diclofenac as DILI in vitromodels. Proteomics analysis of these EVs revealed an increase in enzymes already associated with liver damage, such as catecholamine‐methyl transferase and arginase 1. An increase in translation‐related proteins and a decrease in regulators of apoptosis were also observed. In addition, we show the presence of enzymatic activity of P450 cytochrome 2d1 in EVs. The activity specifically is decreased in EVs secreted by hepatocytes after acetaminophen treatment and increased in EVs derived from GalN‐treated hepatocytes. By using in vivo preclinical models, we demonstrate the presence of this cytochrome activity in circulation under normal conditions and an increased activity after GalN‐induced injury. Conclusion: Hepatocyte‐secreted EVs carry active xenobiotic‐metabolizing enzymes that might be relevant in extracellular metabolism of drugs and be associated with DILI. (Hepatology Communications 2018;0:00‐00)
Collapse
Affiliation(s)
- Laura Palomo
- Exosomes Laboratory, CIC bioGUNE, CIBERehd Bizkaia Spain
| | | | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed Bizkaia Spain
| | | | | | - Felix Elortza
- Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed Bizkaia Spain
| | - Félix Royo
- Exosomes Laboratory, CIC bioGUNE, CIBERehd Bizkaia Spain
| | - Juan M Falcon-Perez
- Exosomes Laboratory, CIC bioGUNE, CIBERehd Bizkaia Spain.,IKERBASQUE, Basque Foundation for Science Bilbao Spain
| |
Collapse
|
36
|
Zhao DS, Jiang LL, Wang LL, Wu ZT, Li ZQ, Shi W, Li P, Jiang Y, Li HJ. Integrated Metabolomics and Proteomics Approach To Identify Metabolic Abnormalities in Rats with Dioscorea bulbifera Rhizome-Induced Hepatotoxicity. Chem Res Toxicol 2018; 31:843-851. [DOI: 10.1021/acs.chemrestox.8b00066] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Dong-Sheng Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Li-Long Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ling-Li Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zi-Tian Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zhuo-Qing Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Wei Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yan Jiang
- Nanjing Forestry University, Nanjing 210037, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| |
Collapse
|
37
|
Saha B, Momen-Heravi F, Furi I, Kodys K, Catalano D, Gangopadhyay A, Haraszti R, Satishchandran A, Iracheta-Vellve A, Adejumo A, Shaffer SA, Szabo G. Extracellular vesicles from mice with alcoholic liver disease carry a distinct protein cargo and induce macrophage activation through heat shock protein 90. Hepatology 2018; 67:1986-2000. [PMID: 29251792 PMCID: PMC5906190 DOI: 10.1002/hep.29732] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/22/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
UNLABELLED A salient feature of alcoholic liver disease (ALD) is Kupffer cell (KC) activation and recruitment of inflammatory monocytes and macrophages (MØs). These key cellular events of ALD pathogenesis may be mediated by extracellular vesicles (EVs). EVs transfer biomaterials, including proteins and microRNAs, and have recently emerged as important effectors of intercellular communication. We hypothesized that circulating EVs from mice with ALD have a protein cargo characteristic of the disease and mediate biological effects by activating immune cells. The total number of circulating EVs was increased in mice with ALD compared to pair-fed controls. Mass spectrometric analysis of circulating EVs revealed a distinct signature for proteins involved in inflammatory responses, cellular development, and cellular movement between ALD EVs and control EVs. We also identified uniquely important proteins in ALD EVs that were not present in control EVs. When ALD EVs were injected intravenously into alcohol-naive mice, we found evidence of uptake of ALD EVs in recipient livers in hepatocytes and MØs. Hepatocytes isolated from mice after transfer of ALD EVs, but not control EVs, showed increased monocyte chemoattractant protein 1 mRNA and protein expression, suggesting a biological effect of ALD EVs. Compared to control EV recipient mice, ALD EV recipient mice had increased numbers of F4/80hi cluster of differentiation 11b (CD11b)lo KCs and increased percentages of tumor necrosis factor alpha-positive/interleukin 12/23-positive (inflammatory/M1) KCs and infiltrating monocytes (F4/80int CD11bhi ), while the percentage of CD206+ CD163+ (anti-inflammatory/M2) KCs was decreased. In vitro, ALD EVs increased tumor necrosis factor alpha and interleukin-1β production in MØs and reduced CD163 and CD206 expression. We identified heat shock protein 90 in ALD EVs as the mediator of ALD-EV-induced MØ activation. CONCLUSION Our study indicates a specific protein signature of ALD EVs and demonstrates a functional role of circulating EVs containing heat shock protein 90 in mediating KC/MØ activation in the liver. (Hepatology 2018;67:1986-2000).
Collapse
Affiliation(s)
- Banishree Saha
- Department of Medicine, University of Massachusetts Medical
School,Department of Polymer Science and Engineering, University of
Massachusetts Amherst
| | - Fatemeh Momen-Heravi
- Department of Medicine, University of Massachusetts Medical
School,College of Dental Medicine, Columbia University
| | - Istvan Furi
- Department of Medicine, University of Massachusetts Medical
School
| | - Karen Kodys
- Department of Medicine, University of Massachusetts Medical
School
| | - Donna Catalano
- Department of Medicine, University of Massachusetts Medical
School
| | | | - Reka Haraszti
- RNA Therapeutics Institute, University of Massachusetts Medical
School
| | | | | | - Adeyinka Adejumo
- Department of Medicine, University of Massachusetts Medical
School,North Shore Medical Center, Salem, MA
| | - Scott A. Shaffer
- Proteomics and Mass Spectrometry Facility and Department of
Biochemistry and Molecular Pharmacology, University of Massachusetts Medical
School
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical
School
| |
Collapse
|
38
|
Yang J, Li C, Zhang L, Wang X. Extracellular Vesicles as Carriers of Non-coding RNAs in Liver Diseases. Front Pharmacol 2018; 9:415. [PMID: 29740327 PMCID: PMC5928552 DOI: 10.3389/fphar.2018.00415] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are small membranous vesicles secreted from normal, diseased, and transformed cells in vitro and in vivo. EVs have been found to play a critical role in cell-to-cell communication by transferring non-coding RNAs (ncRNAs) including microRNAs (miRNAs), long ncRNAs (lncRNAs) and so on. Emerging evidence shows that transferring biological information through EVs to neighboring cells in intercellular communication not only keep physiological functions, but also participate in the pathogenesis of liver diseases. Liver diseases often promote release of EVs and/or in different cargo sorting into these EVs. Either of these modifications can promote disease pathogenesis. Given this fact, EV-associated ncRNAs, such as miR-192, miR-122 and lncRNA-ROR and so on, can serve as new diagnostic biomarkers and new therapeutic targets for liver disease, because altered EV-associated ncRNAs may reflect the underlying liver disease condition. In this review, we focus on understanding the emerging role of EV-associated ncRNAs in viral hepatitis, liver fibrosis, alcoholic hepatitis (AH), non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) and discuss their utility in biomarker discovery and therapeutics. A better understanding of this multifaceted pattern of communication between different type cells in liver may contribute to developing novel approaches for personalized diagnostics and therapeutics.
Collapse
Affiliation(s)
- Junfa Yang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Changyao Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
39
|
Cho YE, Song BJ, Akbar M, Baek MC. Extracellular vesicles as potential biomarkers for alcohol- and drug-induced liver injury and their therapeutic applications. Pharmacol Ther 2018; 187:180-194. [PMID: 29621595 DOI: 10.1016/j.pharmthera.2018.03.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are small membranous vesicles originating from various cells and tissues, including the liver parenchymal hepatocytes and nonparenchymal cells such as Kupffer and stellate cells. Recently, the pathophysiological role of EVs, such as exosomes and microvesicles, has been increasingly recognized based on their properties of intercellular communications. These EVs travel through the circulating blood and interact with specific cells and then deliver their cargos such as nucleic acids and proteins into recipient cells. In addition, based on their stabilities, circulating EVs from body fluids such as blood, cerebrospinal fluid, urine, saliva, semen, breast milk and amniotic fluids are being studied as a valuable source of potential biomarkers for providing information about the physiological status of original cells or tissues. In addition, EVs are considered potential therapeutic agents due to their ability for intercellular communications between different cell types within the liver and between various organs through transfer of their cargos. In this review, we have briefly described recent advances in the characteristics and pathophysiological roles of EVs in alcoholic liver disease (ALD) or drug-induced liver injury (DILI) and discuss their advantages in the discovery of potential biomarkers and therapeutic agents.
Collapse
Affiliation(s)
- Young-Eun Cho
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Mohammed Akbar
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
40
|
Mleczko J, Ortega FJ, Falcon‐Perez JM, Wabitsch M, Fernandez‐Real JM, Mora S. Extracellular Vesicles from Hypoxic Adipocytes and Obese Subjects Reduce Insulin-Stimulated Glucose Uptake. Mol Nutr Food Res 2018; 62:1700917. [PMID: 29292863 PMCID: PMC5887919 DOI: 10.1002/mnfr.201700917] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/14/2017] [Indexed: 12/28/2022]
Abstract
SCOPE We investigate the effects of extracellular vesicles (EVs) obtained from in vitro adipocyte cell models and from obese subjects on glucose transport and insulin responsiveness. METHODS AND RESULTS EVs are isolated from the culture supernatant of adipocytes cultured under normoxia, hypoxia (1% oxygen), or exposed to macrophage conditioned media (15% v/v). EVs are isolated from the plasma of lean individuals and subjects with obesity. Cultured adipocytes are incubated with EVs and activation of insulin signalling cascades and insulin-stimulated glucose transport are measured. EVs released from hypoxic adipocytes impair insulin-stimulated 2-deoxyglucose uptake and reduce insulin mediated phosphorylation of AKT. Insulin-mediated phosphorylation of extracellular regulated kinases (ERK1/2) is not affected. EVs from individuals with obesity decrease insulin stimulated 2-deoxyglucose uptake in adipocytes (p = 0.0159). CONCLUSION EVs released by stressed adipocytes impair insulin action in neighboring adipocytes.
Collapse
Affiliation(s)
- Justyna Mleczko
- Department of Molecular and Cellular PhysiologyInstitute of Translational MedicineThe University of LiverpoolLiverpoolUK
- CIC bioGUNECIBERehdDerioBizkaiaSpain
| | - Francisco J. Ortega
- Department of Molecular and Cellular PhysiologyInstitute of Translational MedicineThe University of LiverpoolLiverpoolUK
- Dept Diabetes Endocrinology and NutritionInstitut Investigacio Biomedica Girona (IdibGI)GironaSpain
- CIBER Obesidad (CIBERObn)Instituto de Salud Carlos IIIMadridSpain
| | | | - Martin Wabitsch
- Division of Pediatric Endocrinology and DiabetesDepartment of Pediatrics and Adolescent MedicineUniversity of UlmGermany
| | - Jose Manuel Fernandez‐Real
- Dept Diabetes Endocrinology and NutritionInstitut Investigacio Biomedica Girona (IdibGI)GironaSpain
- CIBER Obesidad (CIBERObn)Instituto de Salud Carlos IIIMadridSpain
| | - Silvia Mora
- Department of Molecular and Cellular PhysiologyInstitute of Translational MedicineThe University of LiverpoolLiverpoolUK
| |
Collapse
|
41
|
Redox regulation of leukocyte-derived microparticle release and protein content in response to cold physical plasma-derived oxidants. CLINICAL PLASMA MEDICINE 2017. [DOI: 10.1016/j.cpme.2017.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
Szabo G, Momen-Heravi F. Extracellular vesicles in liver disease and potential as biomarkers and therapeutic targets. Nat Rev Gastroenterol Hepatol 2017; 14. [PMID: 28634412 PMCID: PMC6380505 DOI: 10.1038/nrgastro.2017.71] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are membranous vesicles originating from different cells in the liver. The pathophysiological role of EVs is increasingly recognized in liver diseases, including alcoholic liver disease, NAFLD, viral hepatitis and hepatocellular carcinoma. EVs, via their cargo, can provide communication between different cell types in the liver and between organs. EVs are explored as biomarkers of disease and could also represent therapeutic targets and vehicles for treatment delivery. Here, we review advances in understanding the role of EVs in liver diseases and discuss their utility in biomarker discovery and therapeutics.
Collapse
|
43
|
Guo P, Yu H, Wang Y, Xie X, Chen G. Exosome: An Emerging Participant in the Development of Liver Disease. HEPATITIS MONTHLY 2017; 17. [DOI: 10.5812/hepatmon.58021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
44
|
de Castro CA, dos Santos Dias MM, da Silva KA, dos Reis SA, da Conceição LL, De Nadai Marcon L, de Sousa Moraes LF, do Carmo Gouveia Peluzio M. Liver Biomarkers and Their Applications to Nutritional Interventions in Animal Studies. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-94-007-7675-3_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
An M, Lohse I, Tan Z, Zhu J, Wu J, Kurapati H, Morgan MA, Lawrence TS, Cuneo KC, Lubman DM. Quantitative Proteomic Analysis of Serum Exosomes from Patients with Locally Advanced Pancreatic Cancer Undergoing Chemoradiotherapy. J Proteome Res 2017; 16:1763-1772. [PMID: 28240915 DOI: 10.1021/acs.jproteome.7b00024] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer is the third leading cause of cancer-related death in the USA. Despite extensive research, minimal improvements in patient outcomes have been achieved. Early identification of treatment response and metastasis would be valuable to determine the appropriate therapeutic course for patients. In this work, we isolated exosomes from the serum of 10 patients with locally advanced pancreatic cancer at serial time points over a course of therapy, and quantitative analysis was performed using the iTRAQ method. We detected approximately 700-800 exosomal proteins per sample, several of which have been implicated in metastasis and treatment resistance. We compared the exosomal proteome of patients at different time points during treatment to healthy controls and identified eight proteins that show global treatment-specific changes. We then tested the effect of patient-derived exosomes on the migration of tumor cells and found that patient-derived exosomes, but not healthy controls, induce cell migration, supporting their role in metastasis. Our data show that exosomes can be reliably extracted from patient serum and analyzed for protein content. The differential loading of exosomes during a course of therapy suggests that exosomes may provide novel insights into the development of treatment resistance and metastasis.
Collapse
Affiliation(s)
- Mingrui An
- Department of Surgery, University of Michigan Medical Center , Ann Arbor, Michigan 48109, United States
| | - Ines Lohse
- Department of Radiation Oncology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Zhijing Tan
- Department of Surgery, University of Michigan Medical Center , Ann Arbor, Michigan 48109, United States
| | - Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center , Ann Arbor, Michigan 48109, United States
| | - Jing Wu
- Department of Surgery, University of Michigan Medical Center , Ann Arbor, Michigan 48109, United States
| | - Himabindu Kurapati
- Department of Radiation Oncology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Kyle C Cuneo
- Department of Radiation Oncology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - David M Lubman
- Department of Surgery, University of Michigan Medical Center , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
46
|
Jia X, Chen J, Megger DA, Zhang X, Kozlowski M, Zhang L, Fang Z, Li J, Chu Q, Wu M, Li Y, Sitek B, Yuan Z. Label-free Proteomic Analysis of Exosomes Derived from Inducible Hepatitis B Virus-Replicating HepAD38 Cell Line. Mol Cell Proteomics 2017; 16:S144-S160. [PMID: 28242843 DOI: 10.1074/mcp.m116.063503] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 02/17/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major health problem worldwide. Recent evidence suggests that some viruses can manipulate the infection process by packing specific viral and cellular components into exosomes, small nanometer-sized (30-150 nm) vesicles secreted from various cells. However, the impact of HBV replication on the content of exosomes produced by hepatocytes has not been fully delineated. In this work, an HBV-inducible cell line HepAD38 was used to directly compare changes in the protein content of exosomes secreted from HepAD38 cells with or without HBV replication. Exosomes were isolated from supernantants of HepAD38 cells cultured with or without doxycycline (dox) and their purity was confirmed by transmission electron microscopy (TEM) and Western immunoblotting assays. Ion-intensity based label-free LC-MS/MS quantitation technologies were applied to analyze protein content of exosomes from HBV replicating cells [referred as HepAD38 (dox-)-exo] and from HBV nonreplicating cells [referred as HepAD38 (dox+)-exo]. A total of 1412 exosomal protein groups were identified, among which the abundance of 35 proteins was significantly changed following HBV replication. Strikingly, 5 subunit proteins from the 26S proteasome complex, including PSMC1, PSMC2, PSMD1, PSMD7 and PSMD14 were consistently enhanced in HepAD38 (dox-)-exo. Bioinformatic analysis of differential exosomal proteins confirmed the significant enrichment of components involved in the proteasomal catabolic process. Proteasome activity assays further suggested that HepAD38 (dox-)-exo had enhanced proteolytic activity compared with HepAD38 (dox+)-exo. Furthermore, human peripheral monocytes incubated with HepAD38 (dox-)-exo induced a significantly lower level of IL-6 secretion compared with IL-6 levels from HepAD38 (dox+)-exo. Irreversible inhibition of proteasomal activity within exosomes restored higher production of IL-6 by monocytes, suggesting that transmission of proteasome subunit proteins by HepAD38 (dox-)-exo might modulate the production of pro-inflammatory molecules in the recipient monocytes. These results revealed the composition and potential function of exosomes produced during HBV replication, thus providing a new perspective on the role of exosomes in HBV-host interaction.
Collapse
Affiliation(s)
- Xiaofang Jia
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Jieliang Chen
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Dominik A Megger
- §Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Xiaonan Zhang
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Maya Kozlowski
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Lijun Zhang
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Zhong Fang
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Jin Li
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Qiaofang Chu
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Min Wu
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Yaming Li
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Barbara Sitek
- §Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Zhenghong Yuan
- From the ‡Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology, MOE/MOH, Shanghai Medical College, Fudan University, Shanghai 201508, China;
| |
Collapse
|
47
|
Cho YE, Im EJ, Moon PG, Mezey E, Song BJ, Baek MC. Increased liver-specific proteins in circulating extracellular vesicles as potential biomarkers for drug- and alcohol-induced liver injury. PLoS One 2017; 12:e0172463. [PMID: 28225807 PMCID: PMC5321292 DOI: 10.1371/journal.pone.0172463] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/06/2017] [Indexed: 12/21/2022] Open
Abstract
Drug- and alcohol-induced liver injury are a leading cause of liver failure and transplantation. Emerging evidence suggests that extracellular vesicles (EVs) are a source of biomarkers because they contain unique proteins reflecting the identity and tissue-specific origin of the EV proteins. This study aimed to determine whether potentially hepatotoxic agents, such as acetaminophen (APAP) and binge alcohol, can increase the amounts of circulating EVs and evaluate liver-specific EV proteins as potential biomarkers for liver injury. The circulating EVs, isolated from plasma of APAP-exposed, ethanol-fed mice, or alcoholic hepatitis patients versus normal control counterparts, were characterized by proteomics and biochemical methods. Liver specific EV proteins were analyzed by immunoblots and ELISA. The amounts of total and liver-specific proteins in circulating EVs from APAP-treated mice significantly increased in a dose- and time-dependent manner. Proteomic analysis of EVs from APAP-exposed mice revealed that the amounts of liver-specific and/or hepatotoxic proteins were increased compared to those of controls. Additionally, the increased protein amounts in EVs following APAP exposure returned to basal levels when mice were treated with N-acetylcysteine or glutathione. Similar results of increased amounts and liver-specific proteins in circulating EVs were also observed in mice exposed to hepatotoxic doses of thioacetamide or d-galactosamine but not by non-hepatotoxic penicillin or myotoxic bupivacaine. Additionally, binge ethanol exposure significantly elevated liver-specific proteins in circulating EVs from mice and alcoholics with alcoholic hepatitis, compared to control counterparts. These results indicate that circulating EVs in drug- and alcohol-mediated hepatic injury contain liver-specific proteins that could serve as specific biomarkers for hepatotoxicity.
Collapse
Affiliation(s)
- Young-Eun Cho
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, United States of America
| | - Eun-Ju Im
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Pyong-Gon Moon
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Esteban Mezey
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, United States of America
| | - Moon-Chang Baek
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
48
|
Hepatocyte-secreted extracellular vesicles modify blood metabolome and endothelial function by an arginase-dependent mechanism. Sci Rep 2017; 7:42798. [PMID: 28211494 PMCID: PMC5314384 DOI: 10.1038/srep42798] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/13/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes release extracellular vesicles (EVs) loaded with signaling molecules and enzymes into the bloodstream. Although the importance of EVs in the intercellular communication is already recognized, the metabolic impact of the enzymes carried by these vesicles is still unclear. We evaluated the global effect of the enzymatic activities of EVs by performing untargeted metabolomic profiling of serum samples after their exposure to EVs. This approach revealed a significant change in the abundance of 94 serum metabolic signals. Our study shows that these vesicles modify the concentration of metabolites of different chemical nature including metabolites related to arginine metabolism, which regulates vascular function. To assess the functional relevance of this finding, we examined the levels of arginase-1 protein and its activity in the hepatic EVs carrying the exosomal markers CD81 and CD63. Remarkably, the arginase activity was also detected in EVs isolated from the serum in vivo, and this vesicular activity significantly increased under liver-damaging conditions. Finally, we demonstrated that EVs secreted by hepatocytes inhibited the acetylcholine-induced relaxation in isolated pulmonary arteries, via an arginase-dependent mechanism. In summary, our study demonstrates that the hepatocyte-released EVs are metabolically active, affecting a number of serum metabolites involved in oxidative stress metabolism and the endothelial function.
Collapse
|
49
|
Royo F, Palomo L, Mleczko J, Gonzalez E, Alonso C, Martínez I, Pérez-Cormenzana M, Castro A, Falcon-Perez JM. Metabolically active extracellular vesicles released from hepatocytes under drug-induced liver-damaging conditions modify serum metabolome and might affect different pathophysiological processes. Eur J Pharm Sci 2017; 98:51-57. [DOI: 10.1016/j.ejps.2016.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/23/2016] [Accepted: 10/18/2016] [Indexed: 01/06/2023]
|
50
|
Casal E, Palomo L, Cabrera D, Falcon-Perez JM. A Novel Sensitive Method to Measure Catechol-O-Methyltransferase Activity Unravels the Presence of This Activity in Extracellular Vesicles Released by Rat Hepatocytes. Front Pharmacol 2016; 7:501. [PMID: 28066248 PMCID: PMC5179529 DOI: 10.3389/fphar.2016.00501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022] Open
Abstract
There is a clear need for drug treatments to be selected according to the characteristics of an individual patient, in order to improve efficacy and reduce the number and severity of adverse drug reactions. One of the main enzymes to take into account in pharmacogenomics is catechol O-methyltransferase (COMT), which catalyzes the transfer of a methyl group from S-adenosylmethionine to catechols and catecholamines, like the neurotransmitters dopamine, epinephrine, and norepinephrine. Although, most of this enzyme is associated to intracellular vesicles, recently it has also been detected in extracellular vesicles secreted by hepatocytes and in serum circulating vesicles. COMT has implications in many neurological and psychiatric disorders like Parkinson's disease, chronic fatigue, pain response, schizophrenia, and bipolar disorders. Remarkably, genetic variations of COMT affect its activity and are associated to various human disorders from psychiatric diseases to estrogen-induced cancers. Consequently, the establishment of new methods to evaluate COMT activity is an important aspect to investigate the biology of this drug-metabolizing enzyme. Herein, we have developed a sensitive and selective method to determine COMT activity. We first optimized the activity in rat liver incubated with two different substrates; norepinephrine and dopamine. The enzymatically formed products (normetanephrine and 3-methoxytyramine, respectively) were extracted by solid-phase extraction using weak cation exchange cartridges, chromatographically separated, and detected and quantified using a mass spectrometer. The range of quantitation for both products was from 0.005 to 25 μg/mL. This methodology offers acceptable recovery for both enzymatic products (≥75%) and good accuracy and precision (≤15%). The lower limit of quantifications were 0.01 and 0.005 μM for 3-methoxytyramine and normetanephrine, respectively. Importantly, this sensitive assay was able to detect the presence of COMT activity in extracellular vesicles secreted by hepatocytes supporting a potential role of these vesicles in catecholamines and catecholestrogens metabolisms. In addition, the presence of COMT activity in extracellular vesicles opens new possibilities to develop tools to evaluate personalized drug response in a low invasive manner.
Collapse
Affiliation(s)
- Enriqueta Casal
- Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology Park Bizkaia, Spain
| | - Laura Palomo
- Exosomes Laboratory, Metabolomics Unit, CIC bioGUNE, CIBERehd, Bizkaia Technology Park Bizkaia, Spain
| | - Diana Cabrera
- Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology Park Bizkaia, Spain
| | - Juan M Falcon-Perez
- Metabolomics Platform, CIC bioGUNE, CIBERehd, Bizkaia Technology ParkBizkaia, Spain; Exosomes Laboratory, Metabolomics Unit, CIC bioGUNE, CIBERehd, Bizkaia Technology ParkBizkaia, Spain; Ikerbasque, Basque Foundation for ScienceBilbao, Spain
| |
Collapse
|