1
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [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: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Harris DD, Sabe SA, Broadwin M, Stone C, Xu C, Kanuparthy M, Malhotra A, Abid MR, Sellke FW. Intramyocardial injection of hypoxia-conditioned extracellular vesicles increases myocardial perfusion in a swine model of chronic coronary disease. JTCVS OPEN 2024; 20:49-63. [PMID: 39296447 PMCID: PMC11405997 DOI: 10.1016/j.xjon.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 09/21/2024]
Abstract
Objective Coronary artery disease remains a leading cause of morbidity and mortality worldwide. Patients with advanced coronary artery disease who are not eligible for endovascular or surgical revascularization have limited options. Extracellular vesicles have shown potential to improve myocardial function in preclinical models. Extracellular vesicles can be conditioned to modify their components. Hypoxia-conditioned extracellular vesicles have demonstrated the ability to reduce infarct size and apoptosis in small animals. Our objective is to assess the potential benefits of hypoxia-conditioned extracellular vesicles in a large animal model of coronary artery disease. Methods Coronary artery disease was induced in 14 Yorkshire swine by ameroid constriction of the left circumflex coronary artery. Two weeks postsurgery, swine underwent a repeat left thoracotomy for injections of hypoxia-conditioned extracellular vesicles (n = 7) or saline (control, n = 7). Five weeks later, all animals underwent terminal harvest for perfusion measurements and myocardial sectioning. Results Myocardial perfusion analysis demonstrated a trend toward increase at rest and a significant increase during rapid pacing (P = .09, P < .001). There were significant increases in activated phosphorylated endothelial nitric oxide synthase, endothelial nitric oxide synthase, phosphatidylinositol 3-kinase, phosphorylated protein kinase B, and the phosphorylated protein kinase B/protein kinase B ratio in the hypoxia-conditioned extracellular vesicles group compared with the control group (all P < .05). Additionally, there was a significant decrease in the antiangiogenic proteins collagen 18 and angiostatin (P = .01, P = .01) in the hypoxia-conditioned extracellular vesicles group. Conclusions Intramyocardial injection of hypoxia-conditioned extracellular vesicles results in increased myocardial perfusion without a corresponding change in vessel density. Therefore, this improvement in perfusion is possibly due to changes in nitric oxide signaling. Hypoxia-conditioned extracellular vesicles represent a potential therapeutic strategy to increase myocardial perfusion in patients with advanced coronary artery disease.
Collapse
Affiliation(s)
- Dwight D. Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Christopher Stone
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Cynthia Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Meghamsh Kanuparthy
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Akshay Malhotra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
3
|
Harris DD, Sabe SA, Broadwin M, Xu C, Stone C, Kanuparthy M, Malhotra A, Abid MR, Sellke FW. Intramyocardial Injection of Hypoxia-Conditioned Extracellular Vesicles Modulates Response to Oxidative Stress in the Chronically Ischemic Myocardium. Bioengineering (Basel) 2024; 11:125. [PMID: 38391611 PMCID: PMC10886197 DOI: 10.3390/bioengineering11020125] [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: 12/20/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
INTRODUCTION Patients with advanced coronary artery disease (CAD) who are not eligible for stenting or surgical bypass procedures have limited treatment options. Extracellular vesicles (EVs) have emerged as a potential therapeutic target for the treatment of advanced CAD. These EVs can be conditioned to modify their contents. In our previous research, we demonstrated increased perfusion, decreased inflammation, and reduced apoptosis with intramyocardial injection of hypoxia-conditioned EVs (HEVs). The goal of this study is to further understand the function of HEVs by examining their impact on oxidative stress using our clinically relevant and extensively validated swine model of chronic myocardial ischemia. METHODS Fourteen Yorkshire swine underwent a left thoracotomy for the placement of an ameroid constrictor on the left circumflex coronary artery to model chronic myocardial ischemia. After two weeks of recovery, the swine underwent a redo thoracotomy with injection of either HEVs (n = 7) or a saline control (CON, n = 7) into the ischemic myocardium. Five weeks after injection, the swine were subjected to terminal harvest. Protein expression was measured using immunoblotting. OxyBlot analysis and 3-nitrotyrosine staining were used to quantify total oxidative stress. RESULTS There was a significant increase in myocardial expression of the antioxidants SOD 2, GPX-1, HSF-1, UCP-2, catalase, and HO-1 (all p ≤ 0.05) in the HEV group when compared to control animals. The HEVs also exhibited a significant increase in pro-oxidant NADPH oxidase (NOX) 1, NOX 3, p47phox, and p67phox (all p ≤ 0.05). However, no change was observed in the expression of NFkB, KEAP 1, and PRDX1 (all p > 0.05) between the HEV and CON groups. There were no significant differences in total oxidative stress as determined by OxyBlot and 3-nitrotyrosine staining (p = 0.64, p = 0.32) between the groups. CONCLUSIONS Administration of HEVs in ischemic myocardium induces a significant increase in pro- and antioxidant proteins without a net change in total oxidative stress. These findings suggest that HEV-induced changes in redox signaling pathways may play a role in increased perfusion, decreased inflammation, and reduced apoptosis in ischemic myocardium. Further studies are required to determine if HEVs alter the net oxidative stress in ischemic myocardium at an earlier time point of HEV administration.
Collapse
Affiliation(s)
- Dwight D Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Cynthia Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Christopher Stone
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Meghamsh Kanuparthy
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Akshay Malhotra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
4
|
Sabra M, Sabe SA, Harris DD, Xu CM, Broadwin M, Bellam KG, Banerjee D, Abid MR, Sellke FW. Ischemic myocardial inflammatory signaling in starvation versus hypoxia-derived extracellular vesicles: A comparative analysis. JTCVS OPEN 2023; 16:419-428. [PMID: 38204622 PMCID: PMC10775096 DOI: 10.1016/j.xjon.2023.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/17/2023] [Accepted: 10/03/2023] [Indexed: 01/12/2024]
Abstract
Background Coronary artery disease remains a leading cause of death worldwide. Bone mesenchymal stem cell-derived extracellular vesicles (EVs) have shown promise in the setting of myocardial ischemia. Furthermore, the properties of the EVs can be modified via preconditioning of progenitor cells. Previous research from our lab demonstrated a significant decrease in proinflammatory signaling following treatment with EVs derived from starvation preconditioning of human bone mesenchymal stem cells (MVM EVs) in a porcine model of chronic myocardial ischemia. However, rodent models have demonstrated that the use of EVs derived from hypoxia preconditioning of bone mesenchymal stem cells (HYP EVs) may have extended benefits compared to MVM EVs. This study evaluated the effect of HYP EVs on inflammation in a swine model of chronic myocardial ischemia. We hypothesized that HYP EVs would have a greater anti-inflammatory effect than MVM EVs or saline (CON). Methods Yorkshire swine fed a standard diet underwent placement of an ameroid constrictor to the left circumflex artery. Two weeks later, the animals received intramyocardial injection of saline (CON; n = 6), starvation-derived EVs (MVM; n = 10), or hypoxia-derived EVs (HYP; n = 7). After 5 weeks, myocardial perfusion was assessed, and left ventricular myocardial tissue was harvested. Protein expression was measured using immunoblotting. Data were analyzed via the Kruskal-Wallis test or one-way analysis of variance based on the results of a Shapiro-Wilk test. Coronary perfusion was plotted against relative cytokine concentration and analyzed with the Spearman rank-sum test. Results HYP EV treatment was associated with decreased expression of proinflammatory markers interleukin (IL)-6 (P = .03), Pro-IL-1ß (P = .01), IL-17 (P < .01), and NOD-like receptor protein 3 (NLRP3; P < .01) compared to CON. Ischemic tissue from the MVM group showed significantly decreased expression of pro-inflammatory markers NLRP3 (P < .01), IL-17 (P < .01), and HLA class II histocompatibility antigen (P < .01) compared to CON. The MVM group also had decreased expression of anti-inflammatory IL-10 (P = .01) compared to CON counterparts. There were no significant differences in expression of tumor necrosis factor-α, interferon-γ, IL-12, Toll-like receptor-2, and nuclear factor kappa-light-chain-enhancer of activated B cells in either group . There was no correlation between coronary perfusion and cytokine concentration in the MVM or HYP groups, either at rest or with pacing. Conclusions HYP EVs and MVM EVs appear to result in relative decreases in the degree of inflammation in chronically ischemic swine myocardium, independent of coronary perfusion. It is possible that this observed decrease may partially explain the myocardial benefits seen with both HYP and MVM EV treatment.
Collapse
Affiliation(s)
- Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Dwight D. Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Cynthia M. Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Krishnah G. Bellam
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Debolina Banerjee
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
5
|
Harris DD, Sabe SA, Sabra M, Xu CM, Malhotra A, Broadwin M, Banerjee D, Abid MR, Sellke FW. Intramyocardial injection of hypoxia-conditioned extracellular vesicles modulates apoptotic signaling in chronically ischemic myocardium. JTCVS OPEN 2023; 15:220-228. [PMID: 37808040 PMCID: PMC10556811 DOI: 10.1016/j.xjon.2023.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/28/2023] [Accepted: 05/18/2023] [Indexed: 10/10/2023]
Abstract
Objective Limited treatments exist for nonoperative chronic coronary artery disease. Previously, our laboratory has investigated extracellular vesicle (EV) therapy as a potential treatment for chronic coronary artery disease using a swine model and demonstrated improved cardiac function in swine treated with intramyocardial EV injection. Here, we seek to investigate the potential cardiac benefits of EVs by using hypoxia-conditioned EVs (HEV). Specifically, this study aims to investigate the effect of HEV on apoptosis in chronically ischemic myocardium in swine. Methods Fourteen Yorkshire swine underwent placement of an ameroid constrictor on the left circumflex artery. Two weeks later, swine underwent redo left thoracotomy with injection of either saline (control, n = 7) or HEVs (n = 7). After 5 weeks, swine were euthanized for tissue collection. Terminal deoxynucleotidyl transferase dUTP nick end labeling was used to quantify apoptosis. Immunoblotting was used for protein quantification. Results Terminal deoxynucleotidyl transferase dUTP nick end labeling staining showed a decrease in apoptosis in the HEV group compared with the control (P = .049). The HEV group exhibited a significant increase in the anti-apoptotic signaling molecule phospho-BAD (P = .005), a significant decrease in B-cell lymphoma 2 (P = .006) and an increase in the phospho-B-cell lymphoma to B-cell lymphoma 2 ratio (P < .001). Furthermore, the HEV group exhibited increased levels of prosurvival signaling markers including phosphoinositide 3-kinase, phosphor-extracellular signal-regulated kinase 1/2, phospho-forkhead box protein O1, and phospho-protein kinase B to protein kinase B ratio (all P < .05). Conclusions In chronic myocardial ischemia, treatment with HEV results in a decrease in overall apoptosis, possibly through the activation of both pro-survival and anti-apoptotic signaling pathways.
Collapse
Affiliation(s)
- Dwight D. Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Cynthia M. Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Akshay Malhotra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Debolina Banerjee
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - M. Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| |
Collapse
|
6
|
Sareen N, Srivastava A, Alagarsamy KN, Lionetti V, Dhingra S. Stem cells derived exosomes and biomaterials to modulate autophagy and mend broken hearts. Biochim Biophys Acta Mol Basis Dis 2023:166806. [PMID: 37437748 DOI: 10.1016/j.bbadis.2023.166806] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023]
Abstract
Autophagy maintains cellular homeostasis and plays a crucial role in managing pathological conditions including ischemic myocardial injury leading to heart failure (HF). Despite treatments, no intervention can replace lost cardiomyocytes. Stem cell therapy offers potential for post-myocardial infarction repair but struggles with poor cell retention due to immune rejection. In the search for effective therapies, stem cell-derived extracellular vesicles (EVs), especially exosomes, have emerged as promising tools. These tiny bioactive molecule carriers play vital roles in intercellular communication and tissue engineering. They offer numerous therapeutic benefits including modulating immune responses, promoting tissue repair, and boosting angiogenesis. Additionally, biomaterials provide a conducive 3D microenvironment for cell, exosome, and biomolecule delivery, and enhance heart muscle strength, making it a comprehensive cardiac repair strategy. In this regard, the current review delves into the intricate application of extracellular vesicles (EVs) and biomaterials for managing autophagy in the heart muscle during cardiac injury. Central to our investigation is the exploration of how these elements interact within the context of cardiac repair and regeneration. Additionally, this review also casts light on the formidable challenges that plague this field, such as the issues of safety, efficacy, controlled delivery, and acceptance of these therapeutic strategies for effective clinical translation. Addressing these challenges is crucial for unlocking the full therapeutic potential of EV and biomaterial-based therapies and ensuring their successful translation from bench to bedside.
Collapse
Affiliation(s)
- Niketa Sareen
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada; Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | - Abhay Srivastava
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada
| | - Keshav Narayan Alagarsamy
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Rady Faculty of Health Science, University of Manitoba, Winnipeg R2H2A6, MB, Canada.
| |
Collapse
|
7
|
Karnas E, Dudek P, Zuba-Surma EK. Stem cell- derived extracellular vesicles as new tools in regenerative medicine - Immunomodulatory role and future perspectives. Front Immunol 2023; 14:1120175. [PMID: 36761725 PMCID: PMC9902918 DOI: 10.3389/fimmu.2023.1120175] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023] Open
Abstract
In the last few decades, the practical use of stem cells (SCs) in the clinic has attracted significant attention in the regenerative medicine due to the ability of these cells to proliferate and differentiate into other cell types. However, recent findings have demonstrated that the therapeutic capacity of SCs may also be mediated by their ability to secrete biologically active factors, including extracellular vesicles (EVs). Such submicron circular membrane-enveloped vesicles may be released from the cell surface and harbour bioactive cargo in the form of proteins, lipids, mRNA, miRNA, and other regulatory factors. Notably, growing evidence has indicated that EVs may transfer their bioactive content into recipient cells and greatly modulate their functional fate. Thus, they have been recently envisioned as a new class of paracrine factors in cell-to-cell communication. Importantly, EVs may modulate the activity of immune system, playing an important role in the regulation of inflammation, exhibiting broad spectrum of the immunomodulatory activity that promotes the transition from pro-inflammatory to pro-regenerative environment in the site of tissue injury. Consequently, growing interest is placed on attempts to utilize EVs in clinical applications of inflammatory-related dysfunctions as potential next-generation therapeutic factors, alternative to cell-based approaches. In this review we will discuss the current knowledge on the biological properties of SC-derived EVs, with special focus on their role in the regulation of inflammatory response. We will also address recent findings on the immunomodulatory and pro-regenerative activity of EVs in several disease models, including in vitro and in vivo preclinical, as well as clinical studies. Finally, we will highlight the current perspectives and future challenges of emerging EV-based therapeutic strategies of inflammation-related diseases treatment.
Collapse
|
8
|
Aboulgheit A, Karbasiafshar C, Sabra M, Zhang Z, Sodha N, Abid MR, Sellke FW. Extracellular vesicles improve diastolic function and substructure in normal and high-fat diet models of chronic myocardial ischemia. J Thorac Cardiovasc Surg 2022; 164:e371-e384. [PMID: 34756431 PMCID: PMC9005578 DOI: 10.1016/j.jtcvs.2021.07.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/29/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The burden of mortality and morbidity of cardiovascular disease is in part due to substantial fibrosis accelerated by coexisting risk factors. This study aims to evaluate the effect of extracellular vesicle therapy on diastolic function and myocardial fibrosis in the setting of chronic myocardial ischemia with and without a high-fat diet. METHODS Forty male Yorkshire swine were administered a normal or high-fat diet. At 11 weeks of age, they underwent placement of an ameroid constrictor on their left circumflex coronary artery. Both dietary groups then received either intramyocardial injection of vehicle saline as controls or extracellular vesicles as treatment into the ischemic territory (normal diet control, n = 8; high-fat diet controls, n = 11) or extracellular vesicles (normal diet extracellular vesicles, n = 9; high-fat diet extracellular vesicles, n = 12). Five weeks later, hemodynamic parameters, histology, and selected protein expression were evaluated. RESULTS Extracellular vesicles reduced end-diastolic pressure volume relationship (P = .002), perivascular collagen density (P = .031), calcium mineralization (P = .026), and cardiomyocyte diameter (P < .0001), and upregulated osteopontin (P = .0046) and mechanistic target of rapamycin (P = .021). An interaction between extracellular vesicles and diet was observed in the vimentin area (P = .044) and fraction of myofibroblast markers to total vimentin (P = .049). Significant changes across diet were found with reductions in muscle fiber area (P = .026), tumor necrosis factor α (P = .0002), NADPH oxidase 2 and 4 (P = .0036, P = .008), superoxide dismutase 1 (P = .034), and phosphorylated glycogen synthase kinase 3β (P = .020). CONCLUSIONS Extracellular vesicle therapy improved the myocardium's ability to relax and is likely due to structural improvements at the extracellular matrix and cellular levels.
Collapse
Affiliation(s)
- Ahmed Aboulgheit
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | | | - Mohamed Sabra
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI
| | - Zhiqi Zhang
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | - Neel Sodha
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI.
| |
Collapse
|
9
|
Yates AG, Pink RC, Erdbrügger U, Siljander PR, Dellar ER, Pantazi P, Akbar N, Cooke WR, Vatish M, Dias‐Neto E, Anthony DC, Couch Y. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part II: Pathology: Part II: Pathology. J Extracell Vesicles 2022; 11:e12190. [PMID: 35041301 PMCID: PMC8765328 DOI: 10.1002/jev2.12190] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
It is clear from Part I of this series that extracellular vesicles (EVs) play a critical role in maintaining the homeostasis of most, if not all, normal physiological systems. However, the majority of our knowledge about EV signalling has come from studying them in disease. Indeed, EVs have consistently been associated with propagating disease pathophysiology. The analysis of EVs in biofluids, obtained in the clinic, has been an essential of the work to improve our understanding of their role in disease. However, to interfere with EV signalling for therapeutic gain, a more fundamental understanding of the mechanisms by which they contribute to pathogenic processes is required. Only by discovering how the EV populations in different biofluids change-size, number, and physicochemical composition-in clinical samples, may we then begin to unravel their functional roles in translational models in vitro and in vivo, which can then feedback to the clinic. In Part II of this review series, the functional role of EVs in pathology and disease will be discussed, with a focus on in vivo evidence and their potential to be used as both biomarkers and points of therapeutic intervention.
Collapse
Affiliation(s)
- Abi G. Yates
- Department of PharmacologyUniversity of OxfordOxfordUK
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandSt LuciaAustralia
| | - Ryan C. Pink
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Uta Erdbrügger
- Department of Medicine, Division of NephrologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pia R‐M. Siljander
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Elizabeth R. Dellar
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthJohn Radcliffe Hospital, HeadingtonOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthJohn Radcliffe Hospital, HeadingtonOxfordUK
| | - Emmanuel Dias‐Neto
- Laboratory of Medical Genomics. A.C. Camargo Cancer CentreSão PauloBrazil
- Laboratory of Neurosciences (LIM‐27) Institute of PsychiatrySão Paulo Medical SchoolSão PauloBrazil
| | | | - Yvonne Couch
- Acute Stroke Programme ‐ Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
10
|
Ma S, Yan J, Chen L, Zhu Y, Chen K, Zheng C, Shen M, Liao Y. A Bibliometric and Visualized Analysis of Cardiac Regeneration Over a 20-Year Period. Front Cardiovasc Med 2021; 8:789503. [PMID: 34966800 PMCID: PMC8710530 DOI: 10.3389/fcvm.2021.789503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/01/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Recent research has suggested that cardiac regeneration may have the widely applicable potential of treating heart failure (HF). A comprehensive understanding of the development status of this field is conducive to its development. However, no bibliometric analysis has summarized this field properly. We aimed to analyze cardiac regeneration-related literature over 20 years and provide valuable insights. Methods: Publications were collected from the Web of Science Core Collection (WoSCC). Microsoft Excel, VOSviewer, CiteSpace, and alluvial generator were used to analyze and present the data. Results: The collected 11,700 publications showed an annually increasing trend. The United States and Harvard University were the leading force among all the countries and institutions. The majority of articles were published in Circulation Research, and Circulation was the most co-cited journal. According to co-citation analysis, burst detection and alluvial flow map, cardiomyocyte proliferation, stem cells, such as first-and second-generation, extracellular vesicles especially exosomes, direct cardiac reprogramming, macrophages, microRNAs, and inflammation have become more and more popular recently. Conclusions: Cardiac regeneration remains a research hotspot and develops rapidly. How to modify cardiac regeneration endogenously and exogenously may still be the hotspot in the future and should be discussed more deeply.
Collapse
Affiliation(s)
- Siyuan Ma
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junyu Yan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lu Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingqi Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaitong Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cankun Zheng
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengjia Shen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Shock and Microcirculation, Department of Cardiology, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
van de Wakker SI, van Oudheusden J, Mol EA, Roefs MT, Zheng W, Görgens A, El Andaloussi S, Sluijter JPG, Vader P. Influence of short term storage conditions, concentration methodsand excipients on extracellular vesicle recovery and function. Eur J Pharm Biopharm 2021; 170:59-69. [PMID: 34864197 DOI: 10.1016/j.ejpb.2021.11.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/16/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022]
Abstract
Extracellular vesicles (EVs) are phospholipid bilayer enclosed vesicles which play an important role in intercellular communication. To date, many studies have focused on therapeutic application of EVs. However, to progress EV applications faster towards the clinic, more information about the physical stability and scalable production of EVs is needed. The goal of this study was to evaluate EV recovery and function after varying several conditions in the isolation process or during storage. Physical stability and recovery rates of EVs were evaluated by measuring EV size, particle and protein yields using nanoparticle tracking analysis, microBCA protein quantification assay and transmission electron microscopy. Western blot analyses of specific EV markers were performed to determine EV yields and purity. EV functionality was tested in an endothelial cell wound healing assay. Higher EV recovery rates were found when using HEPES buffered saline (HBS) as buffer compared to phosphate buffered saline (PBS) during EV isolation. When concentrating EVs, 15 ml spinfilters with a 10 kDa membrane cutoff gave the highest EV recovery. Next, EV storage in polypropylene tubes was shown to be superior compared to glass tubes. The use of protective excipients during EV storage, i.e. bovine serum albumin (BSA) and Tween 20, improved EV preservation without influencing their functionality. Finally, it was shown that both 4 °C and -80 °C are suitable for short term storage of EVs. Together, our results indicate that optimizing buffer compositions, concentrating steps, protective excipients and storage properties may collectively increase EV recovery rates significantly while preserving their functional properties, which accelerates translation of EV-based therapeutics towards clinical application.
Collapse
Affiliation(s)
- S I van de Wakker
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - J van Oudheusden
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - E A Mol
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - M T Roefs
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - W Zheng
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - A Görgens
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - S El Andaloussi
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Stockholm, Sweden
| | - J P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - P Vader
- Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht University, the Netherlands; CDL Research, University Medical Center Utrecht, the Netherlands.
| |
Collapse
|
12
|
Salybekov AA, Salybekova A, Sheng Y, Shinozaki Y, Yokoyama K, Kobayashi S, Asahara T. Extracellular Vesicles Derived From Regeneration Associated Cells Preserve Heart Function After Ischemia-Induced Injury. Front Cardiovasc Med 2021; 8:754254. [PMID: 34746267 PMCID: PMC8564358 DOI: 10.3389/fcvm.2021.754254] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
Under vasculogenic conditioning, pro-inflammatory cell subsets of peripheral blood mononuclear cells (PBMCs) shift their phenotype to pro-regenerative cells such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells, collectively designated as regeneration-associated cells (RACs). In this study, we evaluated the therapeutic efficacy of RAC-derived extracellular vesicles (RACev) compared to mesenchymal stem cell-derived EVs (MSCev) in the context of myocardial ischemia reperfusion injury (M-IRI). Human PBMCs were cultured with defined growth factors for seven days to harvest RACs. RACev and MSCev were isolated via serial centrifugation and ultracentrifugation. EV quantity and size were characterized by nanoparticle tracking analysis. In vitro, RACev markedly enhanced the viability, and proliferation of human umbilical vein endothelial cells in a dose-dependent manner compared to MSCev. Notably, systemic injection of RACev improved cardiac functions at 4 weeks, such as fractional shortening, and protection from mitral regurgitation than the MSCev-treated group. Histologically, the RACev-transplanted group showed less interstitial fibrosis and enhanced capillary densities compared to the MSCev group. These beneficial effects were coupled with significant expression of angiogenesis, anti-fibrosis, anti-inflammatory, and cardiomyogenesis-related miRs in RACev, while modestly in MSCev. In vivo bioluminescence analysis showed preferential accumulation of RACev in the IR-injured myocardium, while MSCev accumulation was limited. Immune phenotyping analysis confirmed the immunomodulatory effect of MSCev and RACev. Overall, repetitive systemic transplantation of RACev is superior to MSCev in terms of cardiac function enhancements via crucial angiogenesis, anti-fibrosis, anti-inflammation miR delivery to the ischemic tissue.
Collapse
Affiliation(s)
- Amankeldi A Salybekov
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan.,Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Japan.,Division of Regenerative Medicine, Department of Center for Clinical and Translational Science, Shonan Kamakura General Hospital, Kamakura, Japan.,Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Ainur Salybekova
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Yin Sheng
- Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - Yoshiko Shinozaki
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan
| | - Keiko Yokoyama
- Teaching and Research Support Core Center, Tokai University School of Medicine, Isehara, Japan
| | - Shuzo Kobayashi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Japan.,Division of Regenerative Medicine, Department of Center for Clinical and Translational Science, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Japan.,Department of Advanced Medicine Science, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
13
|
Han C, Yang J, Sun J, Qin G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol Ther 2021; 233:108025. [PMID: 34687770 PMCID: PMC9018895 DOI: 10.1016/j.pharmthera.2021.108025] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/15/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are lipid bilayer particles naturally released from the cell. While exosomes are formed as intraluminal vesicles (ILVs) of the multivesicular endosomes (MVEs) and released to extracellular space upon MVE-plasma membrane fusion, microvesicles are generated through direct outward budding of the plasma membrane. Exosomes and microvesicles have same membrane orientation, different yet overlapping sizes; their cargo contents are selectively packed and dependent on the source cell type and functional state. Both exosomes and microvesicles can transfer bioactive RNAs, proteins, lipids, and metabolites from donor to recipient cells and influence the biological properties of the latter. Over the last decade, their potential roles as effective inter-tissue communicators in cardiovascular physiology and pathology have been increasingly appreciated. In addition, EVs are attractive sources of biomarkers for the diagnosis and prognosis of diseases, because they acquire their complex cargoes through cellular processes intimately linked to disease pathogenesis. Furthermore, EVs obtained from various stem/progenitor cell populations have been tested as cell-free therapy in various preclinical models of cardiovascular diseases and demonstrate unequivocally encouraging benefits. Here we summarize the findings from recent research on the biological functions of EVs in the ischemic heart disease and heart failure, and their potential as novel diagnostic biomarkers and therapeutic opportunities.
Collapse
Affiliation(s)
- Chaoshan Han
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Junjie Yang
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Jiacheng Sun
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL 35294, USA.
| |
Collapse
|
14
|
Yeganeh A, Alibhai FJ, Tobin SW, Lim F, Wu J, Li S, Weisel RD, Li RK. Age-related defects in autophagy alter the secretion of paracrine factors from bone marrow mononuclear cells. Aging (Albany NY) 2021; 13:14687-14708. [PMID: 34088884 PMCID: PMC8221303 DOI: 10.18632/aging.203127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/14/2021] [Indexed: 01/08/2023]
Abstract
Bone marrow mononuclear cell therapy improves cardiac repair after myocardial infarction (MI), in-part through signaling to resident cardiac cells, such as fibroblasts, which regulate scar formation. The efficacy of cell therapy declines with age, as aging of both donor and recipient cells decreases repair responses. Autophagy regulates the microenvironment by both extracellular vesicle (EV)-dependent and independent secretion pathways. We hypothesized that age-related autophagy changes in bone marrow cells (BMCs) alter paracrine signaling, contributing to lower cell therapy efficacy. Here, we demonstrate that young Sca-1+ BMCs exhibited a higher LC3II/LC3I ratio compared to old Sca-1+ BMCs, which was accentuated when BMCs were cultured under hypoxia. To examine the effect on paracrine signaling, old cardiac fibroblasts were cultured with conditioned medium (CM) from young and old Sca-1+ BMCs. Young, but not old CM, enhanced fibroblast proliferation, migration, and differentiation, plus reduced senescence. These beneficial effects were lost when autophagy or EV secretion in BMCs was blocked pharmacologically, or by siRNA knockdown of Atg7. Therefore, both EV-dependent and -independent paracrine signaling from young BMCs is responsible for paracrine stimulation of old cardiac fibroblasts. In vivo, bone marrow chimerism of old mice with young BMCs increased the number of LC3b+ cells in the heart compared to old mice reconstituted with old BMCs. These data suggest that the deterioration of autophagy with aging negatively impacts the paracrine effects of BMCs, and provide mechanistic insight into the age-related decline in cell therapy efficacy that could be targeted to improve the function of old donor cells.
Collapse
Affiliation(s)
- Azadeh Yeganeh
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Faisal J. Alibhai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Stephanie W. Tobin
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Fievel Lim
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Shuhong Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Richard D. Weisel
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, Canada
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
15
|
Karbasiafshar C, Sellke FW, Abid MR. Mesenchymal stem cell-derived extracellular vesicles in the failing heart: past, present, and future. Am J Physiol Heart Circ Physiol 2021; 320:H1999-H2010. [PMID: 33861149 PMCID: PMC8163643 DOI: 10.1152/ajpheart.00951.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. Current treatment options include lifestyle changes, medication, and surgical intervention. However, many patients are unsuitable candidates for surgeries due to comorbidities, diffuse coronary artery disease, or advanced stages of heart failure. The search for new treatment options has recently transitioned from cell-based therapies to stem-cell-derived extracellular vesicles (EVs). A number of challenges remain in the EV field, including the effect of comorbidities, characterization, and delivery. However, recent revolutionary developments and insight into the potential of personalizing EV contents by bioengineering methods to alter specific signaling pathways in the ischemic myocardium hold promise. Here, we discuss the past limitations of cell-based therapies and recent EV studies involving in vivo, in vitro, and omics, and future challenges and opportunities in EV-based treatments in CVD.
Collapse
Affiliation(s)
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Department of Surgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Department of Surgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
16
|
Sabra M, Karbasiafshar C, Aboulgheit A, Raj S, Abid MR, Sellke FW. Clinical Application of Novel Therapies for Coronary Angiogenesis: Overview, Challenges, and Prospects. Int J Mol Sci 2021; 22:3722. [PMID: 33918396 PMCID: PMC8038234 DOI: 10.3390/ijms22073722] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 01/26/2023] Open
Abstract
Cardiovascular diseases continue to be the leading cause of death worldwide, with ischemic heart disease as the most significant contributor. Pharmacological and surgical interventions have improved clinical outcomes, but are unable to ameliorate advanced stages of end-heart failure. Successful preclinical studies of new therapeutic modalities aimed at revascularization have shown short lasting to no effects in the clinical practice. This lack of success may be attributed to current challenges in patient selection, endpoint measurements, comorbidities, and delivery systems. Although challenges remain, the field of therapeutic angiogenesis is evolving, as novel strategies and bioengineering approaches emerge to optimize delivery and efficacy. Here, we describe the structure, vascularization, and regulation of the vascular system with particular attention to the endothelium. We proceed to discuss preclinical and clinical findings and present challenges and future prospects in the field.
Collapse
Affiliation(s)
- Mohamed Sabra
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA; (M.S.); (C.K.); (A.A.); ; (M.R.A.)
| | - Catherine Karbasiafshar
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA; (M.S.); (C.K.); (A.A.); ; (M.R.A.)
| | - Ahmed Aboulgheit
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA; (M.S.); (C.K.); (A.A.); ; (M.R.A.)
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Sidharth Raj
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - M. Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA; (M.S.); (C.K.); (A.A.); ; (M.R.A.)
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Frank W. Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA; (M.S.); (C.K.); (A.A.); ; (M.R.A.)
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Providence, RI 02903, USA;
| |
Collapse
|
17
|
Murali VP, Holmes CA. Biomaterial-based extracellular vesicle delivery for therapeutic applications. Acta Biomater 2021; 124:88-107. [PMID: 33454381 DOI: 10.1016/j.actbio.2021.01.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicle (EV)- based therapies have been successfully tested in preclinical models for several biomedical applications, including tissue engineering, drug delivery and cancer therapy. However, EVs are most commonly delivered via local or systemic injection, which results in rapid clearance. In order to prolong the retention of EVs at target site and improve their therapeutic efficacy, biomaterial-based delivery systems are being investigated. This review discusses the various biomaterial-based systems that have been used to deliver EVs for therapeutic applications, specifically highlighting any strategies for controlled release. Further, challenges to clinical translation of biomaterial-based EV delivery systems are also discussed.
Collapse
Affiliation(s)
- Vishnu Priya Murali
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA.
| | - Christina A Holmes
- Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, 2525 Pottsdamer Street, Room A131, Tallahassee, FL 32310, USA.
| |
Collapse
|
18
|
Lin Y, Anderson JD, Rahnama LMA, Gu SV, Knowlton AA. Exosomes in disease and regeneration: biological functions, diagnostics, and beneficial effects. Am J Physiol Heart Circ Physiol 2020; 319:H1162-H1180. [PMID: 32986962 PMCID: PMC7792703 DOI: 10.1152/ajpheart.00075.2020] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/30/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Exosomes are a subtype of extracellular vesicles. They range from 30 to 150 nm in diameter and originate from intraluminal vesicles. Exosomes were first identified as the mechanism for releasing unnecessary molecules from reticulocytes as they matured to red blood cells. Since then, exosomes have been shown to be secreted by a broad spectrum of cells and play an important role in the cardiovascular system. Different stimuli are associated with increased exosome release and result in different exosome content. The release of harmful DNA and other molecules via exosomes has been proposed as a mechanism to maintain cellular homeostasis. Because exosomes contain parent cell-specific proteins on the membrane and in the cargo that is delivered to recipient cells, exosomes are potential diagnostic biomarkers of various types of diseases, including cardiovascular disease. As exosomes are readily taken up by other cells, stem cell-derived exosomes have been recognized as a potential cell-free regenerative therapy to repair not only the injured heart but other tissues as well. The objective of this review is to provide an overview of the biological functions of exosomes in heart disease and tissue regeneration. Therefore, state-of-the-art methods for exosome isolation and characterization, as well as approaches to assess exosome functional properties, are reviewed. Investigation of exosomes provides a new approach to the study of disease and biological processes. Exosomes provide a potential "liquid biopsy," as they are present in most, if not all, biological fluids that are released by a wide range of cell types.
Collapse
Affiliation(s)
- Yun Lin
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | | | - Lily M A Rahnama
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | - Shenwen V Gu
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | - Anne A Knowlton
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| |
Collapse
|
19
|
Alibhai FJ, Li RK. Understanding systemic factors in aging and rejuvenation. Aging (Albany NY) 2020; 12:20936-20937. [PMID: 33216730 PMCID: PMC7695419 DOI: 10.18632/aging.104213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Faisal J Alibhai
- Toronto General Hospital Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Ontario, Canada.,Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Abdollahi S. Extracellular vesicles from organoids and 3D culture systems. Biotechnol Bioeng 2020; 118:1029-1049. [PMID: 33085083 DOI: 10.1002/bit.27606] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/17/2020] [Accepted: 10/09/2020] [Indexed: 12/28/2022]
Abstract
When discovered, extracellular vesicles (EVs) such as exosomes were thought of as junk carriers and a means by which the cell disposed of its waste material. Over the years, the role of EVs in cell communication has become apparent with the discovery that the nano-scale vesicles also transport RNA, DNA, and other bioactive components to and from the cells. These findings were originally made in EVs from body fluids of organisms and from in vitro two-dimensional (2D) cell culture models. Recently, organoids and other 3D multicellular in vitro models are being used to study EVs in the context of both physiologic and pathological states. However, standard, reproducible methods are lacking for EV analysis using these models. As a step toward understanding the implications of these platforms, this review provides a comprehensive picture of the progress using 3D in vitro culture models for EV analysis. Translational efforts and regulatory considerations for EV therapeutics are also briefly overviewed to understand what is needed for scale-up and, ultimately, commercialization.
Collapse
Affiliation(s)
- Sara Abdollahi
- Department of Human Genetics, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Recent Advances in Understandings Towards Pathogenesis and Treatment for Intrauterine Adhesion and Disruptive Insights from Single-Cell Analysis. Reprod Sci 2020; 28:1812-1826. [PMID: 33125685 PMCID: PMC8189970 DOI: 10.1007/s43032-020-00343-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
Intrauterine adhesion is a major cause of menstrual irregularities, infertility, and recurrent pregnancy losses and the progress towards its amelioration and therapy is slow and unsatisfactory. We aim to summarize and evaluate the current treatment progress and research methods for intrauterine adhesion. We conducted literature review in January 2020 by searching articles at PubMed on prevention and treatment, pathogenesis, the repair of other tissues/organs, cell plasticity, and the stem cell–related therapies for intrauterine adhesion. A total of 110 articles were selected for review. Uterine cell heterogeneity, expression profile, and cell-cell interaction were investigated based on scRNA-seq of uterus provided by Human Cell Landscape (HCL) project. Previous knowledge on intrauterine adhesion (IUA) pathogenesis was mostly derived from correlation studies by differentially expressed genes between endometrial tissue of intrauterine adhesion patients/animal models and normal endometrial tissue. Although the TGF-β1/SMAD pathway was suggested as the key driver for IUA pathogenesis, uterine cell heterogeneity and distinct expression profile among different cell types highlighted the importance of single-cell investigations. Cell-cell interaction in the uterus revealed the central hub of endothelial cells interacting with other cells, with endothelial cells in endothelial to mesenchymal transition and fibroblasts as the strongest interaction partners. The potential of stem cell–related therapies appeared promising, yet suffers from largely animal studies and nonstandard study design. The need to dissect the roles of endometrial cells, endothelial cells, and fibroblasts and their interaction is evident in order to elucidate the molecular and cellular mechanisms in both intrauterine adhesion pathogenesis and treatment.
Collapse
|
22
|
Tobin SW, Alibhai FJ, Weisel RD, Li RK. Considering Cause and Effect of Immune Cell Aging on Cardiac Repair after Myocardial Infarction. Cells 2020; 9:E1894. [PMID: 32823583 PMCID: PMC7465938 DOI: 10.3390/cells9081894] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
The importance of the immune system for cardiac repair following myocardial infarction is undeniable; however, the complex nature of immune cell behavior has limited the ability to develop effective therapeutics. This limitation highlights the need for a better understanding of the function of each immune cell population during the inflammatory and resolution phases of cardiac repair. The development of reliable therapies is further complicated by aging, which is associated with a decline in cell and organ function and the onset of cardiovascular and immunological diseases. Aging of the immune system has important consequences on heart function as both chronic cardiac inflammation and an impaired immune response to cardiac injury are observed in older individuals. Several studies have suggested that rejuvenating the aged immune system may be a valid therapeutic candidate to prevent or treat heart disease. Here, we review the basic patterns of immune cell behavior after myocardial infarction and discuss the autonomous and nonautonomous manners of hematopoietic stem cell and immune cell aging. Lastly, we identify prospective therapies that may rejuvenate the aged immune system to improve heart function such as anti-inflammatory and senolytic therapies, bone marrow transplant, niche remodeling and regulation of immune cell differentiation.
Collapse
Affiliation(s)
- Stephanie W. Tobin
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Faisal J. Alibhai
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
| | - Richard D. Weisel
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5T 1P5, Canada; (S.W.T.); (F.J.A.); (R.D.W.)
- Division of Cardiac Surgery, Peter Munk Cardiac Centre, Toronto General Hospital and University of Toronto, Toronto, ON M5G 2N2, Canada
| |
Collapse
|
23
|
Rogers RG, Ciullo A, Marbán E, Ibrahim AG. Extracellular Vesicles as Therapeutic Agents for Cardiac Fibrosis. Front Physiol 2020; 11:479. [PMID: 32528309 PMCID: PMC7255103 DOI: 10.3389/fphys.2020.00479] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022] Open
Abstract
Heart disease remains an increasing major public health challenge in the United States and worldwide. A common end-organ feature in diseased hearts is myocardial fibrosis, which stiffens the heart and interferes with normal pump function, leading to pump failure. The development of cells for regenerative therapy has been met with many pitfalls on its path to clinical translation. Recognizing that regenerative cells secrete therapeutically bioactive vesicles has paved the way to circumvent many failures of cell therapy. In this review, we provide an overview of extracellular vesicles (EVs), with a focus on their utility as therapeutic agents for cardiac regeneration. We also highlight the engineering potential of EVs to enhance their therapeutic application.
Collapse
Affiliation(s)
| | | | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | |
Collapse
|
24
|
Maghin E, Garbati P, Quarto R, Piccoli M, Bollini S. Young at Heart: Combining Strategies to Rejuvenate Endogenous Mechanisms of Cardiac Repair. Front Bioeng Biotechnol 2020; 8:447. [PMID: 32478060 PMCID: PMC7237726 DOI: 10.3389/fbioe.2020.00447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
True cardiac regeneration of the injured heart has been broadly described in lower vertebrates by active replacement of lost cardiomyocytes to functionally and structurally restore the myocardial tissue. On the contrary, following severe injury (i.e., myocardial infarction) the adult mammalian heart is endowed with an impaired reparative response by means of meager wound healing program and detrimental remodeling, which can lead over time to cardiomyopathy and heart failure. Lately, a growing body of basic, translational and clinical studies have supported the therapeutic use of stem cells to provide myocardial regeneration, with the working hypothesis that stem cells delivered to the cardiac tissue could result into new cardiovascular cells to replenish the lost ones. Nevertheless, multiple independent evidences have demonstrated that injected stem cells are more likely to modulate the cardiac tissue via beneficial paracrine effects, which can enhance cardiac repair and reinstate the embryonic program and cell cycle activity of endogenous cardiac stromal cells and resident cardiomyocytes. Therefore, increasing interest has been addressed to the therapeutic profiling of the stem cell-derived secretome (namely the total of cell-secreted soluble factors), with specific attention to cell-released extracellular vesicles, including exosomes, carrying cardioprotective and regenerative RNA molecules. In addition, the use of cardiac decellularized extracellular matrix has been recently suggested as promising biomaterial to develop novel therapeutic strategies for myocardial repair, as either source of molecular cues for regeneration, biological scaffold for cardiac tissue engineering or biomaterial platform for the functional release of factors. In this review, we will specifically address the translational relevance of these two approaches with ad hoc interest in their feasibility to rejuvenate endogenous mechanisms of cardiac repair up to functional regeneration.
Collapse
Affiliation(s)
- Edoardo Maghin
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.,Department of Women's and Children Health, University of Padova, Padua, Italy
| | - Patrizia Garbati
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Rodolfo Quarto
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy.,UOC Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Martina Piccoli
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
25
|
Ozaki Tan SJ, Floriano JF, Nicastro L, Emanueli C, Catapano F. Novel Applications of Mesenchymal Stem Cell-derived Exosomes for Myocardial Infarction Therapeutics. Biomolecules 2020; 10:E707. [PMID: 32370160 PMCID: PMC7277090 DOI: 10.3390/biom10050707] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and morbidity globally, representing approximately a third of all deaths every year. The greater part of these cases is represented by myocardial infarction (MI), or heart attack as it is better known, which occurs when declining blood flow to the heart causes injury to cardiac tissue. Mesenchymal stem cells (MSCs) are multipotent stem cells that represent a promising vector for cell therapies that aim to treat MI due to their potent regenerative effects. However, it remains unclear the extent to which MSC-based therapies are able to induce regeneration in the heart and even less clear the degree to which clinical outcomes could be improved. Exosomes, which are small extracellular vesicles (EVs) known to have implications in intracellular communication, derived from MSCs (MSC-Exos), have recently emerged as a novel cell-free vector that is capable of conferring cardio-protection and regeneration in target cardiac cells. In this review, we assess the current state of research of MSC-Exos in the context of MI. In particular, we place emphasis on the mechanisms of action by which MSC-Exos accomplish their therapeutic effects, along with commentary on the current difficulties faced with exosome research and the ongoing clinical applications of stem-cell derived exosomes in different medical contexts.
Collapse
Affiliation(s)
- Sho Joseph Ozaki Tan
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Juliana Ferreria Floriano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
- Botucatu Medical School, Sao Paulo State University, Botucatu 18618687, Brazil
| | - Laura Nicastro
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Francesco Catapano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| |
Collapse
|
26
|
Saxena S, Kumar S. Pharmacotherapy to gene editing: potential therapeutic approaches for Hutchinson-Gilford progeria syndrome. GeroScience 2020; 42:467-494. [PMID: 32048129 PMCID: PMC7205988 DOI: 10.1007/s11357-020-00167-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), commonly called progeria, is an extremely rare disorder that affects only one child per four million births. It is characterized by accelerated aging in affected individuals leading to premature death at an average age of 14.5 years due to cardiovascular complications. The main cause of HGPS is a sporadic autosomal dominant point mutation in LMNA gene resulting in differently spliced lamin A protein known as progerin. Accumulation of progerin under nuclear lamina and activation of its downstream effectors cause perturbation in cellular morphology and physiology which leads to a systemic disorder that mainly impairs the cardiovascular system, bones, skin, and overall growth. Till now, no cure has been found for this catastrophic disorder; however, several therapeutic strategies are under development. The current review focuses on the overall progress in the field of therapeutic approaches for the management/cure of HGPS. We have also discussed the new disease models that have been developed for the study of this rare disorder. Moreover, we have highlighted the therapeutic application of extracellular vesicles derived from stem cells against aging and aging-related disorders and, therefore, suggest the same for the treatment of HGPS.
Collapse
Affiliation(s)
- Saurabh Saxena
- Department of Medical Laboratory Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India.
| | - Sanjeev Kumar
- Faculty of Technology and Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| |
Collapse
|
27
|
Alibhai FJ, Lim F, Yeganeh A, DiStefano PV, Binesh‐Marvasti T, Belfiore A, Wlodarek L, Gustafson D, Millar S, Li S, Weisel RD, Fish JE, Li R. Cellular senescence contributes to age-dependent changes in circulating extracellular vesicle cargo and function. Aging Cell 2020; 19:e13103. [PMID: 31960578 PMCID: PMC7059145 DOI: 10.1111/acel.13103] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 11/20/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have emerged as important regulators of inter‐cellular and inter‐organ communication, in part via the transfer of their cargo to recipient cells. Although circulating EVs have been previously studied as biomarkers of aging, how circulating EVs change with age and the underlying mechanisms that contribute to these changes are poorly understood. Here, we demonstrate that aging has a profound effect on the circulating EV pool, as evidenced by changes in concentration, size, and cargo. Aging also alters particle function; treatment of cells with EV fractions isolated from old plasma reduces macrophage responses to lipopolysaccharide, increases phagocytosis, and reduces endothelial cell responses to vascular endothelial growth factor compared to cells treated with young EV fractions. Depletion studies indicate that CD63+ particles mediate these effects. Treatment of macrophages with EV‐like particles revealed that old particles increased the expression of EV miRNAs in recipient cells. Transfection of cells with microRNA mimics recapitulated some of the effects seen with old EV‐like particles. Investigation into the underlying mechanisms using bone marrow transplant studies revealed circulating cell age does not substantially affect the expression of aging‐associated circulating EV miRNAs in old mice. Instead, we show that cellular senescence contributes to changes in particle cargo and function. Notably, senolytic treatment of old mice shifted plasma particle cargo and function toward that of a younger phenotype. Collectively, these results demonstrate that senescent cells contribute to changes in plasma EVs with age and suggest a new mechanism by which senescent cells can affect cellular functions throughout the body.
Collapse
Affiliation(s)
- Faisal J. Alibhai
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Fievel Lim
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Azadeh Yeganeh
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Peter V. DiStefano
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Tina Binesh‐Marvasti
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Alyssa Belfiore
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Lukasz Wlodarek
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Dakota Gustafson
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto ON Canada
| | - Sean Millar
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Shu‐Hong Li
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
| | - Richard D. Weisel
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
- Division of Cardiac Surgery Peter Munk Cardiac CentreToronto General Hospital and University of Toronto Toronto ON Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
- Department of Laboratory Medicine and Pathobiology University of Toronto Toronto ON Canada
| | - Ren‐Ke Li
- Toronto General Hospital Research Institute Toronto General Hospital Toronto ON Canada
- Division of Cardiac Surgery Peter Munk Cardiac CentreToronto General Hospital and University of Toronto Toronto ON Canada
| |
Collapse
|
28
|
Thankam FG, Agrawal DK. Infarct Zone: a Novel Platform for Exosome Trade in Cardiac Tissue Regeneration. J Cardiovasc Transl Res 2020; 13:686-701. [PMID: 31907784 DOI: 10.1007/s12265-019-09952-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022]
Abstract
The global incidence of coronary artery diseases (CADs), especially myocardial infarction (MI), has drastically increased in recent years. Even though the conventional therapies have improved the outcomes, the post-MI complications and the increased rate of recurrence among the survivors are still alarming. Molecular events associated with the pathogenesis and the adaptive responses of the surviving myocardium are largely unknown. Focus on exosome-mediated signaling for cell-cell/matrix communications at the infarct zone reflects an emerging opportunity in cardiac regeneration. Also, cardiac tissue engineering provides promising insights for the next generation of therapeutic approaches in the management of CADs. In this article, we critically reviewed the current understanding on the biology of cardiac exosomes, therapeutic potential of exosomes, and recent developments in cardiac tissue engineering and discussed novel translational approaches based on tissue engineering and exosomes for cardiac regeneration and CADs.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
29
|
Leitolis A, Suss PH, Roderjan JG, Angulski ABB, da Costa FDA, Stimamiglio MA, Correa A. Human Heart Explant-Derived Extracellular Vesicles: Characterization and Effects on the In Vitro Recellularization of Decellularized Heart Valves. Int J Mol Sci 2019; 20:ijms20061279. [PMID: 30875722 PMCID: PMC6471048 DOI: 10.3390/ijms20061279] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are particles released from different cell types and represent key components of paracrine secretion. Accumulating evidence supports the beneficial effects of EVs for tissue regeneration. In this study, discarded human heart tissues were used to isolate human heart-derived extracellular vesicles (hH-EVs). We used nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM) to physically characterize hH-EVs and mass spectrometry (MS) to profile the protein content in these particles. The MS analysis identified a total of 1248 proteins. Gene ontology (GO) enrichment analysis in hH-EVs revealed the proteins involved in processes, such as the regulation of cell death and response to wounding. The potential of hH-EVs to induce proliferation, adhesion, angiogenesis and wound healing was investigated in vitro. Our findings demonstrate that hH-EVs have the potential to induce proliferation and angiogenesis in endothelial cells, improve wound healing and reduce mesenchymal stem-cell adhesion. Last, we showed that hH-EVs were able to significantly promote mesenchymal stem-cell recellularization of decellularized porcine heart valve leaflets. Altogether our data confirmed that hH-EVs modulate cellular processes, shedding light on the potential of these particles for tissue regeneration and for scaffold recellularization.
Collapse
Affiliation(s)
- Amanda Leitolis
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Paraná, Curitiba 81350-010, Brazil.
| | - Paula Hansen Suss
- Pontifical Catholic University of Paraná-PUCPR, Curitiba 80215-901, Brazil.
| | | | - Addeli Bez Batti Angulski
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Paraná, Curitiba 81350-010, Brazil.
| | | | - Marco Augusto Stimamiglio
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Paraná, Curitiba 81350-010, Brazil.
| | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Paraná, Curitiba 81350-010, Brazil.
| |
Collapse
|
30
|
Alibhai FJ, Li RK. Commentary: Circulating factors released after myocardial infarction: Beneficial or detrimental? J Thorac Cardiovasc Surg 2018; 157:2270-2271. [PMID: 30503734 DOI: 10.1016/j.jtcvs.2018.10.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Faisal J Alibhai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|