1
|
Farahzadi R, Fathi E, Valipour B, Ghaffary S. Stem cells-derived exosomes as cardiac regenerative agents. IJC HEART & VASCULATURE 2024; 52:101399. [PMID: 38584674 PMCID: PMC10990901 DOI: 10.1016/j.ijcha.2024.101399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/03/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
Heart failure is a root cause of morbidity and mortality worldwide. Due to the limited regenerative capacity of the heart following myocardial injury, stem cell-based therapies have been considered a hopeful approach for improving cardiac regeneration. In recent years, different kinds of cell products have been investigated regarding their potential to treat patients with heart failure. Despite special attention to cell therapy and its products, therapeutic efficacy has been disappointing, and clinical application is not affordable. In the past few years, a subset of small extracellular vehicles (EVs), commonly known as "exosomes," was reported to grant regenerative and cardioprotective signals at a value similar to their donor cells. The conceptual advantage is that they may be ideally used without evoking a relevant recipient immune response or other adverse effects associated with viable cells. The evidence related to their beneficial effects in animal models of heart failure is rapidly growing. However, there is remarkable heterogeneity regarding source cells, isolation process, effective dosage, and delivery mode. This brief review will focus on the latest research and debates on regenerative potential and cardiac repair of exosomes from different sources, such as cardiac/non-cardiac stem, somatic cells, and progenitor cells. Overall, the current state of research on exosomes as an experimental therapy for heart diseases will be discussed.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Ghaffary
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Olejarz W, Sadowski K, Radoszkiewicz K. Extracellular Vesicles in Atherosclerosis: State of the Art. Int J Mol Sci 2023; 25:388. [PMID: 38203558 PMCID: PMC10779125 DOI: 10.3390/ijms25010388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/17/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease driven by lipid accumulation in the arteries, leading to narrowing and thrombosis that causes mortality. Emerging evidence has confirmed that atherosclerosis affects younger people and is involved in the majority of deaths worldwide. EVs are associated with critical steps in atherosclerosis, cholesterol metabolism, immune response, endothelial dysfunction, vascular inflammation, and remodeling. Endothelial cell-derived EVs can interact with platelets and monocytes, thereby influencing endothelial dysfunction, atherosclerotic plaque destabilization, and the formation of thrombus. EVs are potential diagnostic and prognostic biomarkers in atherosclerosis (AS) and cardiovascular disease (CVD). Importantly, EVs derived from stem/progenitor cells are essential mediators of cardiogenesis and cardioprotection and may be used in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| |
Collapse
|
3
|
Ghassemi K, Inouye K, Takhmazyan T, Bonavida V, Yang JW, de Barros NR, Thankam FG. Engineered Vesicles and Hydrogel Technologies for Myocardial Regeneration. Gels 2023; 9:824. [PMID: 37888397 PMCID: PMC10606880 DOI: 10.3390/gels9100824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Increased prevalence of cardiovascular disease and potentially life-threatening complications of myocardial infarction (MI) has led to emerging therapeutic approaches focusing on myocardial regeneration and restoration of physiologic function following infarction. Extracellular vesicle (EV) technology has gained attention owing to the biological potential to modulate cellular immune responses and promote the repair of damaged tissue. Also, EVs are involved in local and distant cellular communication following damage and play an important role in initiating the repair process. Vesicles derived from stem cells and cardiomyocytes (CM) are of particular interest due to their ability to promote cell growth, proliferation, and angiogenesis following MI. Although a promising candidate for myocardial repair, EV technology is limited by the short retention time of vesicles and rapid elimination by the body. There have been several successful attempts to address this shortcoming, which includes hydrogel technology for the sustained bioavailability of EVs. This review discusses and summarizes current understanding regarding EV technology in the context of myocardial repair.
Collapse
Affiliation(s)
- Kaitlyn Ghassemi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Keiko Inouye
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Tatevik Takhmazyan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Victor Bonavida
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; (J.-W.Y.); (N.R.d.B.)
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; (J.-W.Y.); (N.R.d.B.)
| | - Finosh G. Thankam
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| |
Collapse
|
4
|
Li G, Chen T, Dahlman J, Eniola‐Adefeso L, Ghiran IC, Kurre P, Lam WA, Lang JK, Marbán E, Martín P, Momma S, Moos M, Nelson DJ, Raffai RL, Ren X, Sluijter JPG, Stott SL, Vunjak‐Novakovic G, Walker ND, Wang Z, Witwer KW, Yang PC, Lundberg MS, Ochocinska MJ, Wong R, Zhou G, Chan SY, Das S, Sundd P. Current challenges and future directions for engineering extracellular vesicles for heart, lung, blood and sleep diseases. J Extracell Vesicles 2023; 12:e12305. [PMID: 36775986 PMCID: PMC9923045 DOI: 10.1002/jev2.12305] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/19/2022] [Accepted: 01/09/2022] [Indexed: 02/14/2023] Open
Abstract
Extracellular vesicles (EVs) carry diverse bioactive components including nucleic acids, proteins, lipids and metabolites that play versatile roles in intercellular and interorgan communication. The capability to modulate their stability, tissue-specific targeting and cargo render EVs as promising nanotherapeutics for treating heart, lung, blood and sleep (HLBS) diseases. However, current limitations in large-scale manufacturing of therapeutic-grade EVs, and knowledge gaps in EV biogenesis and heterogeneity pose significant challenges in their clinical application as diagnostics or therapeutics for HLBS diseases. To address these challenges, a strategic workshop with multidisciplinary experts in EV biology and U.S. Food and Drug Administration (USFDA) officials was convened by the National Heart, Lung and Blood Institute. The presentations and discussions were focused on summarizing the current state of science and technology for engineering therapeutic EVs for HLBS diseases, identifying critical knowledge gaps and regulatory challenges and suggesting potential solutions to promulgate translation of therapeutic EVs to the clinic. Benchmarks to meet the critical quality attributes set by the USFDA for other cell-based therapeutics were discussed. Development of novel strategies and approaches for scaling-up EV production and the quality control/quality analysis (QC/QA) of EV-based therapeutics were recognized as the necessary milestones for future investigations.
Collapse
Affiliation(s)
- Guoping Li
- Cardiovascular Research CenterMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Tianji Chen
- Department of Pediatrics, College of MedicineUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - James Dahlman
- Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGeorgiaUSA
| | - Lola Eniola‐Adefeso
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Ionita C. Ghiran
- Department of Anesthesia and Pain MedicineBeth Israel Deaconess Medical Center, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Peter Kurre
- Children's Hospital of Philadelphia, Comprehensive Bone Marrow Failure Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Department of PediatricsEmory School of MedicineAflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University and Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Jennifer K. Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical SciencesVeterans Affairs Western New York Healthcare SystemBuffaloNew YorkUSA
| | - Eduardo Marbán
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Stefan Momma
- Institute of Neurology (Edinger Institute)University HospitalGoethe UniversityFrankfurt am MainGermany
| | - Malcolm Moos
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Deborah J. Nelson
- Department of Pharmacological and Physiological SciencesThe University of ChicagoChicagoIllinoisUSA
| | - Robert L. Raffai
- Department of Veterans Affairs, Surgical Service (112G)San Francisco VA Medical CenterSan FranciscoCaliforniaUSA
- Department of Surgery, Division of Vascular and Endovascular SurgeryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Xi Ren
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Joost P. G. Sluijter
- Department of Experimental Cardiology, Circulatory Health LaboratoryRegenerative Medicine Centre, UMC Utrecht, University UtrechtUtrechtThe Netherlands
| | - Shannon L. Stott
- Massachusetts General Hospital Cancer Center and Harvard Medical SchoolBostonMassachusettsUSA
| | - Gordana Vunjak‐Novakovic
- Department of Biomedical Engineering, Department of MedicineColumbia UniversityNew YorkNew YorkUSA
| | - Nykia D. Walker
- Department of Biological SciencesUniversity of Maryland Baltimore CountyBaltimoreMarylandUSA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Department of Neurology and Neurosurgeryand The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Phillip C. Yang
- Division of Cardiovascular Medicine, Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Martha S. Lundberg
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Margaret J. Ochocinska
- Division of Blood Diseases and Resources, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Renee Wong
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Guofei Zhou
- Division of Lung Diseases, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung and Blood Vascular Medicine InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of Cardiology and Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Saumya Das
- Cardiovascular Research CenterMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Prithu Sundd
- Pittsburgh Heart, Lung and Blood Vascular Medicine InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of Pulmonary Allergy and Critical Care Medicine and Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
5
|
An Z, Tian J, Liu Y, Zhao X, Yang X, Yong J, Liu L, Zhang L, Jiang W, Song X, Zhang H. Exosomes as a Cell-free Therapy for Myocardial Injury Following Acute Myocardial Infarction or Ischemic Reperfusion. Aging Dis 2022; 13:1770-1786. [PMID: 36465167 PMCID: PMC9662265 DOI: 10.14336/ad.2022.0416] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/16/2022] [Indexed: 08/13/2023] Open
Abstract
Exosomes, which contain miRNA, have been receiving growing attention in cardiovascular therapy because of their role in mediating cell-cell communication, autophagy, apoptosis, inflammation, and angiogenesis. Several studies have suggested that miRNA derived from exosomes can be used to detect myocardial infarctions (MI) in patients. Basic research also suggests that exosomes could serve as a potential therapeutic target for treating acute myocardial infarction. Ischemia/reperfusion (IR) injury is associated with adverse cardiac events after acute MI. We aim to review the potential benefits and mechanisms of exosomes in treating MI and IR injury.
Collapse
Affiliation(s)
- Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Yue Liu
- Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jingwen Yong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Lijun Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Liu N, Xie L, Xiao P, Chen X, Kong W, Lou Q, Chen F, Lu X. Cardiac fibroblasts secrete exosome microRNA to suppress cardiomyocyte pyroptosis in myocardial ischemia/reperfusion injury. Mol Cell Biochem 2022; 477:1249-1260. [PMID: 35119583 PMCID: PMC8913441 DOI: 10.1007/s11010-021-04343-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/22/2021] [Indexed: 01/30/2023]
Abstract
Molecular mechanisms underlying myocardial ischemia/reperfusion (MI/R) injury and effective strategies to treat MI/R injury are both in shortage. Although pyroptosis of cardiomyocytes and the protective role of cardiac fibroblasts (CFs) have been well recognized as targets to reduce MI/R injury and sudden cardiac death (SCD), the connection has not yet been established. Here, we showed that CFs protected cardiomyocytes against MI/R-induced injury through suppression of pyroptosis. A novel molecular mechanism underpinning this effect was further identified. Under hypoxia/reoxygenation condition, CFs were found to secrete exosomes, which contain increased level of microRNA-133a (miR-133a). These exosomes then delivered miR-133a into cardiomyocytes to target ELAVL1 and repressed cardiomyocyte pyroptosis. Based on this finding, we successfully developed a new strategy that used exosomes derived from CFs with overexpressed miR-133a to enhance the therapeutic outcomes for the MI/R injury. Overall, our results provide a novel molecular basis for understanding and treating MI/R injury, and our study also provides novel insight for the postmortem diagnosis of MI/R injury induced SCD by using exosome biomarker in forensic.
Collapse
Affiliation(s)
- Niannian Liu
- Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China.,Department of Forensic Medicine, Nanjing Medical University, No. 101 Longmian Road, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Liang Xie
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210000, Jiangsu, China
| | - Pingxi Xiao
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, 211166, China
| | - Xing Chen
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, 211166, China
| | - Wenjie Kong
- Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China
| | - Qiaozhen Lou
- Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, China
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, No. 101 Longmian Road, Jiangning District, Nanjing, 211166, Jiangsu, China.
| | - Xiang Lu
- Department of Geriatrics, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
7
|
Lodrini AM, Goumans MJ. Cardiomyocytes Cellular Phenotypes After Myocardial Infarction. Front Cardiovasc Med 2021; 8:750510. [PMID: 34820429 PMCID: PMC8606669 DOI: 10.3389/fcvm.2021.750510] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
Despite the increasing success of interventional coronary reperfusion strategies, mortality related to acute myocardial infarction (MI) is still substantial. MI is defined as sudden death of myocardial tissue caused by an ischemic episode. Ischaemia leads to adverse remodelling in the affected myocardium, inducing metabolic and ionic perturbations at a single cell level, ultimately leading to cell death. The adult mammalian heart has limited regenerative capacity to replace lost cells. Identifying and enhancing physiological cardioprotective processes may be a promising therapy for patients with MI. Studies report an increasing amount of evidence stating the intricacy of the pathophysiology of the infarcted heart. Besides apoptosis, other cellular phenotypes have emerged as key players in the ischemic myocardium, in particular senescence, inflammation, and dedifferentiation. Furthermore, some cardiomyocytes in the infarct border zone uncouple from the surviving myocardium and dedifferentiate, while other cells become senescent in response to injury and start to produce a pro-inflammatory secretome. Enhancing electric coupling between cardiomyocytes in the border zone, eliminating senescent cells with senolytic compounds, and upregulating cardioprotective cellular processes like autophagy, may increase the number of functional cardiomyocytes and therefore enhance cardiac contractility. This review describes the different cellular phenotypes and pathways implicated in injury, remodelling, and regeneration of the myocardium after MI. Moreover, we discuss implications of the complex pathophysiological attributes of the infarcted heart in designing new therapeutic strategies.
Collapse
Affiliation(s)
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
8
|
Zeng CY, Xu J, Liu X, Lu YQ. Cardioprotective Roles of Endothelial Progenitor Cell-Derived Exosomes. Front Cardiovasc Med 2021; 8:717536. [PMID: 34513956 PMCID: PMC8428070 DOI: 10.3389/fcvm.2021.717536] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
With the globally increasing prevalence, cardiovascular diseases (CVDs) have become the leading cause of mortality. The transplantation of endothelial progenitor cells (EPCs) holds a great promise due to their potential for vasculogenesis, angiogenesis, and protective cytokine release, whose mechanisms are essential for CVD therapies. In reality, many investigations have attributed the therapeutic effects of EPC transplantation to the secretion of paracrine factors rather than the differentiation function. Of note, previous studies have suggested that EPCs could also release exosomes (diameter range of 30–150 nm), which carry various lipids and proteins and are abundant in microRNAs. The EPC-derived exosomes (EPC-EXs) were reported to act on the heart and blood vessels and were implicated in anti-inflammation, anti-oxidation, anti-apoptosis, the inhibition of endothelial-to-mesenchymal transition (EndMT), and cardiac fibrosis, as well as anti-vascular remodeling and angiogenesis, which were considered as protective effects against CVDs. In this review, we summarize the current knowledge on using EPC-EXs as therapeutic agents and provide a detailed description of their identified mechanisms of action to promote the prognosis of CVDs.
Collapse
Affiliation(s)
- Cai-Yu Zeng
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jia Xu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xin Liu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Qiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Exosomal miR-218-5p/miR-363-3p from Endothelial Progenitor Cells Ameliorate Myocardial Infarction by Targeting the p53/JMY Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5529430. [PMID: 34326916 PMCID: PMC8302385 DOI: 10.1155/2021/5529430] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/08/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022]
Abstract
Accumulating evidence has shown that endothelial progenitor cell-derived exosomes (EPC-Exos) can ameliorate myocardial fibrosis. The purpose of the present study was to investigate the effects of EPC-Exos-derived microRNAs (miRNAs) on myocardial infarction (MI). A miRNA-Seq dataset of miRNAs differentially expressed between EPCs and exosomes was collected. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to validate the miRNA expression indicated by miRNA-Seq. Immunofluorescence, cell proliferation, and angiogenesis assays were employed to investigate the effects of miRNAs on cardiac fibroblasts (CFs) in vitro. Interactions between miRNAs and their respective targets were examined via immunoblotting, qRT-PCR, and luciferase reporter assays. An MI rat model was constructed, and various staining and immunohistochemical assays were performed to explore the mechanisms underlying the miRNA-mediated effects on MI. miR-363-3p and miR-218-5p were enriched in EPC-Exos, and miR-218-5p and miR-363-3p mimic or inhibitor enhanced or suppressed CF proliferation and angiogenesis, respectively. miR-218-5p and miR-363-3p regulated p53 and junction-mediating and regulatory protein (JMY) by binding to the promoter region of p53 and the 3′ untranslated region of JMY. Additionally, treatment of CFs with Exo-miR-218-5p or Exo-miR-363-3p upregulated p53 and downregulated JMY expression, promoted mesenchymal-endothelial transition, and inhibited myocardial fibrosis. Administration of exosomes containing miR-218-5p mimic or miR-363-3p mimic ameliorated left coronary artery ligation-induced MI and restored myocardial tissue integrity in the MI model rats. In summary, these results show that the protective ability of EPC-Exos against MI was mediated by the shuttled miR-218-5p or miR-363-3p via targeting of the p53/JMY signaling pathway.
Collapse
|
10
|
Santoso MR, Ikeda G, Tada Y, Jung JH, Vaskova E, Sierra RG, Gati C, Goldstone AB, von Bornstaedt D, Shukla P, Wu JC, Wakatsuki S, Woo YJ, Yang PC. Exosomes From Induced Pluripotent Stem Cell-Derived Cardiomyocytes Promote Autophagy for Myocardial Repair. J Am Heart Assoc 2020; 9:e014345. [PMID: 32131688 PMCID: PMC7335524 DOI: 10.1161/jaha.119.014345] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Induced pluripotent stem cells and their differentiated cardiomyocytes (iCMs) have tremendous potential as patient‐specific therapy for ischemic cardiomyopathy following myocardial infarctions, but difficulties in viable transplantation limit clinical translation. Exosomes secreted from iCMs (iCM‐Ex) can be robustly collected in vitro and injected in lieu of live iCMs as a cell‐free therapy for myocardial infarction. Methods and Results iCM‐Ex were precipitated from iCM supernatant and characterized by protein marker expression, nanoparticle tracking analysis, and functionalized nanogold transmission electron microscopy. iCM‐Ex were then used in in vitro and in vivo models of ischemic injuries. Cardiac function in vivo was evaluated by left ventricular ejection fraction and myocardial viability measurements by magnetic resonance imaging. Cardioprotective mechanisms were studied by JC‐1 (tetraethylbenzimidazolylcarbocyanine iodide) assay, immunohistochemistry, quantitative real‐time polymerase chain reaction, transmission electron microscopy, and immunoblotting. iCM‐Ex measured ≈140 nm and expressed CD63 and CD9. iCM and iCM‐Ex microRNA profiles had significant overlap, indicating that exosomal content was reflective of the parent cell. Mice treated with iCM‐Ex demonstrated significant cardiac improvement post–myocardial infarction, with significantly reduced apoptosis and fibrosis. In vitro iCM apoptosis was significantly reduced by hypoxia and exosome biogenesis inhibition and restored by treatment with iCM‐Ex or rapamycin. Autophagosome production and autophagy flux was upregulated in iCM‐Ex groups in vivo and in vitro. Conclusions iCM‐Ex improve post–myocardial infarction cardiac function by regulating autophagy in hypoxic cardiomyoytes, enabling a cell‐free, patient‐specific therapy for ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Michelle R Santoso
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Gentaro Ikeda
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Yuko Tada
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Ji-Hye Jung
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Evgeniya Vaskova
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Raymond G Sierra
- Hard X-ray Department LCLS SLAC National Accelerator Laboratory Menlo Park CA.,Stanford PULSE Institute SLAC National Accelerator Laboratory Menlo Park CA
| | - Cornelius Gati
- Department of Structural Biology Stanford University School of Medicine Stanford CA
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery Falk Building Stanford University Medical Center Stanford CA
| | | | - Praveen Shukla
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Joseph C Wu
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| | - Soichi Wakatsuki
- Department of Structural Biology Stanford University School of Medicine Stanford CA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery Falk Building Stanford University Medical Center Stanford CA
| | - Phillip C Yang
- Stanford Cardiovascular Institute Stanford University School of Medicine Stanford CA
| |
Collapse
|
11
|
Santos MF, Rappa G, Karbanová J, Vanier C, Morimoto C, Corbeil D, Lorico A. Anti-human CD9 antibody Fab fragment impairs the internalization of extracellular vesicles and the nuclear transfer of their cargo proteins. J Cell Mol Med 2019; 23:4408-4421. [PMID: 30982221 PMCID: PMC6533511 DOI: 10.1111/jcmm.14334] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/28/2019] [Accepted: 03/21/2019] [Indexed: 12/14/2022] Open
Abstract
The intercellular communication mediated by extracellular vesicles (EVs) has gained international interest during the last decade. Interfering with the mechanisms regulating this cellular process might find application particularly in oncology where cancer cell‐derived EVs play a role in tumour microenvironment transformation. Although several mechanisms were ascribed to explain the internalization of EVs, little is our knowledge about the fate of their cargos, which are crucial to mediate their function. We recently demonstrated a new intracellular pathway in which a fraction of endocytosed EV‐associated proteins is transported into the nucleoplasm of the host cell via a subpopulation of late endosomes penetrating into the nucleoplasmic reticulum. Silencing tetraspanin CD9 both in EVs and recipient cells strongly decreased the endocytosis of EVs and abolished the nuclear transfer of their cargos. Here, we investigated whether monovalent Fab fragments derived from 5H9 anti‐CD9 monoclonal antibody (referred hereafter as CD9 Fab) interfered with these cellular processes. To monitor the intracellular transport of proteins, we used fluorescent EVs containing CD9‐green fluorescent protein fusion protein and various melanoma cell lines and bone marrow‐derived mesenchymal stromal cells as recipient cells. Interestingly, CD9 Fab considerably reduced EV uptake and the nuclear transfer of their proteins in all examined cells. In contrast, the divalent CD9 antibody stimulated both events. By impeding intercellular communication in the tumour microenvironment, CD9 Fab‐mediated inhibition of EV uptake, combined with direct targeting of cancerous cells could lead to the development of novel anti‐melanoma therapeutic strategies.
Collapse
Affiliation(s)
- Mark F Santos
- College of Medicine, Touro University Nevada, Henderson, Nevada
| | - Germana Rappa
- College of Medicine, Touro University Nevada, Henderson, Nevada
| | - Jana Karbanová
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Cheryl Vanier
- College of Medicine, Touro University Nevada, Henderson, Nevada
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Denis Corbeil
- Biotechnology Center and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Aurelio Lorico
- College of Medicine, Touro University Nevada, Henderson, Nevada.,Mediterranean Institute of Oncology, Viagrande, Italy
| |
Collapse
|
12
|
Hocine HR, Brunel S, Chen Q, Giustiniani J, San Roman MJ, Ferrat YJ, Palacios I, de la Rosa O, Lombardo E, Bensussan A, Charron D, Jabrane-Ferrat N, Al-Daccak R. Extracellular Vesicles Released by Allogeneic Human Cardiac Stem/Progenitor Cells as Part of Their Therapeutic Benefit. Stem Cells Transl Med 2019; 8:911-924. [PMID: 30924311 PMCID: PMC6708067 DOI: 10.1002/sctm.18-0256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/18/2019] [Indexed: 12/28/2022] Open
Abstract
The positive effects of therapeutic human allogeneic cardiac stem/progenitor cells (hCPC) in terms of cardiac repair/regeneration are very likely mediated by paracrine effects. Our previous studies revealed the advantageous immune interactions of allogeneic hCPC and proposed them as part of the positive paracrine effects occurring upon their application postmyocardial infarction (MI). Currently, extracellular vesicles/exosomes (EV/Exs) released by stem/progenitor cells are also proposed as major mediators of paracrine effects of therapeutic cells. Along this line, we evaluated contribution of EV/Exs released by therapeutic hCPC to the benefit of their successful allogeneic clinical application. Through tailored allogeneic in vitro human assay models mimicking the clinical setting, we demonstrate that hCPC‐released EV/Exs were rapidly and efficiently up‐taken by chief cellular actors of cardiac repair/regeneration. This promoted MAPK/Erk1/2 activation, migration, and proliferation of human leukocyte antigens (HLA)‐mismatched hCPC, mimicking endogenous progenitor cells and cardiomyocytes, and enhanced endothelial cell migration, growth, and organization into tube‐like structures through activation of several signaling pathways. EV/Exs also acted as pro‐survival stimuli for HLA‐mismatched monocytes tuning their phenotype toward an intermediate anti‐inflammatory pro‐angiogenic phenotype. Thus, while positively impacting the intrinsic regenerative and angiogenic programs, EV/Exs released by therapeutic allogeneic hCPC can also actively contribute to shaping MI‐inflammatory environment, which could strengthen the benefits of hCPC allogeneic interactions. Collectively, our data might forecast the application of allogeneic hCPC followed by their cell‐free EV/Exs as a strategy that will not only elicit the cell‐contact mediated reparative/regenerative immune response but also have the desired long‐lasting effects through the EV/Exs. stem cells translational medicine2019;8:911&924
Collapse
Affiliation(s)
- Hocine Rachid Hocine
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-976, Université Paris-Diderot, Hôpital Saint-Louis, Paris, France.,HLA et Médecine, Labex Transplantex, Hôpital Saint Louis, Paris, France
| | - Simon Brunel
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-976, Université Paris-Diderot, Hôpital Saint-Louis, Paris, France
| | - Qian Chen
- Centre of Pathophysiology Toulouse Purpan, INSERM U1043, CNRS UMR5282, Toulouse III University, Toulouse, France
| | - Jerome Giustiniani
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-976, Université Paris-Diderot, Hôpital Saint-Louis, Paris, France.,Institut Jean Godinot, Unicancer, Reims, France
| | | | - Yann J Ferrat
- CERAG Laboratory, University of Grenoble Alpes, Grenoble, France
| | | | | | | | - Armand Bensussan
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-976, Université Paris-Diderot, Hôpital Saint-Louis, Paris, France
| | - Dominique Charron
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-976, Université Paris-Diderot, Hôpital Saint-Louis, Paris, France.,HLA et Médecine, Labex Transplantex, Hôpital Saint Louis, Paris, France
| | - Nabila Jabrane-Ferrat
- Centre of Pathophysiology Toulouse Purpan, INSERM U1043, CNRS UMR5282, Toulouse III University, Toulouse, France
| | - Reem Al-Daccak
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS-976, Université Paris-Diderot, Hôpital Saint-Louis, Paris, France.,HLA et Médecine, Labex Transplantex, Hôpital Saint Louis, Paris, France
| |
Collapse
|
13
|
Yuan KM, Zhang PH, Qi SS, Zhu QZ, Li P. Emerging Role for Exosomes in the Progress of Stem Cell Research. Am J Med Sci 2018; 356:481-486. [DOI: 10.1016/j.amjms.2018.07.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 06/27/2018] [Accepted: 07/13/2018] [Indexed: 01/08/2023]
|
14
|
Lazar E, Benedek T, Korodi S, Rat N, Lo J, Benedek I. Stem cell-derived exosomes - an emerging tool for myocardial regeneration. World J Stem Cells 2018; 10:106-115. [PMID: 30190780 PMCID: PMC6121000 DOI: 10.4252/wjsc.v10.i8.106] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs) continue to represent the number one cause of death and disability in industrialized countries. The most severe form of CVD is acute myocardial infarction (AMI), a devastating disease associated with high mortality and disability. In a substantial proportion of patients who survive AMI, loss of functional cardiomyocytes as a result of ischaemic injury leads to ventricular failure, resulting in significant alteration to quality of life and increased mortality. Therefore, many attempts have been made in recent years to identify new tools for the regeneration of functional cardiomyocytes. Regenerative therapy currently represents the ultimate goal for restoring the function of damaged myocardium by stimulating the regeneration of the infarcted tissue or by providing cells that can generate new myocardial tissue to replace the damaged tissue. Stem cells (SCs) have been proposed as a viable therapy option in these cases. However, despite the great enthusiasm at the beginning of the SC era, justified by promising initial results, this therapy has failed to demonstrate a significant benefit in large clinical trials. One interesting finding of SC studies is that exosomes released by mesenchymal SCs (MSCs) are able to enhance the viability of cardiomyocytes after ischaemia/reperfusion injury, suggesting that the beneficial effects of MSCs in the recovery of functional myocardium could be related to their capacity to secrete exosomes. Ten years ago, it was discovered that exosomes have the unique property of transferring miRNA between cells, acting as miRNA nanocarriers. Therefore, exosome-based therapy has recently been proposed as an emerging tool for cardiac regeneration as an alternative to SC therapy in the post-infarction period. This review aims to discuss the emerging role of exosomes in developing innovative therapies for cardiac regeneration as well as their potential role as candidate biomarkers or for developing new diagnostic tools.
Collapse
Affiliation(s)
- Erzsebet Lazar
- Department of Internal Medicine, Clinic of Haematology and Bone Marrow Transplantation, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540042, Romania
| | - Theodora Benedek
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| | - Szilamer Korodi
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| | - Nora Rat
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| | - Jocelyn Lo
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, United States
| | - Imre Benedek
- Department of Internal Medicine, Clinic of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Tirgu Mures 540136, Romania
- Department of Advanced Research in Multimodality Cardiac Imaging, Cardio Med Medical Center, Tirgu Mures 540124, Romania
| |
Collapse
|