1
|
Fatehi Hassanabad A, Zarzycki AN, Patel VB, Fedak PWM. Current concepts in the epigenetic regulation of cardiac fibrosis. Cardiovasc Pathol 2024; 73:107673. [PMID: 38996851 DOI: 10.1016/j.carpath.2024.107673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
Cardiac fibrosis is a significant driver of congestive heart failure, a syndrome that continues to affect a growing patient population globally. Cardiac fibrosis results from a constellation of complex processes at the transcription, receptor, and signaling axes levels. Various mediators and signaling cascades, such as the transformation growth factor-beta pathway, have been implicated in the pathophysiology of cardiac tissue fibrosis. Our understanding of these markers and pathways has improved in recent years as more advanced technologies and assays have been developed, allowing for better delineation of the crosstalk between specific factors. There is mounting evidence suggesting that epigenetic modulation plays a pivotal role in the progression of cardiac fibrosis. Transcriptional regulation of key pro- and antifibrotic pathways can accentuate or blunt the rate and extent of fibrosis at the tissue level. Exosomes, micro-RNAs, and long noncoding RNAs all belong to factors that can impact the epigenetic signature in cardiac fibrosis. Herein, we comprehensively review the latest literature about exosomes, their contents, and cardiac fibrosis. In doing so, we highlight the specific transcriptional factors with pro- or antifibrotic properties. We also assimilate the data supporting these mediators' potential utility as diagnostic or prognostic biomarkers. Finally, we offer insight into where further work can be done to fill existing gaps to translate preclinical findings better and improve clinical outcomes.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
2
|
Moreno A, Alarcón-Zapata P, Guzmán-Gútierrez E, Radojkovic C, Contreras H, Nova-Lampeti E, A Zúñiga F, Rodriguez-Alvárez L, Escudero C, Lagos P, Aguayo C. Changes in the Release of Endothelial Extracellular Vesicles CD144+, CCR6+, and CXCR3+ in Individuals with Acute Myocardial Infarction. Biomedicines 2024; 12:2119. [PMID: 39335632 PMCID: PMC11430588 DOI: 10.3390/biomedicines12092119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Acute myocardial infarction (AMI) results from vulnerable plaque rupture, causing ischemic cardiomyocyte necrosis and intense inflammation. Paradoxically, this inflammation releases factors that aid heart repair. Recent findings suggest a role for extracellular vesicles (EVs) in intercellular communication during post-AMI cardiac repair. However, EVs' tissue origin and chemokine profile in the blood of patients with AMI remains unclear. This study characterized the tissue origin and chemokine receptor profile of EVs in the coronary and peripheral blood of patients with AMI. The results reveal that vesicles isolated from coronary and peripheral blood plasma are enriched in tetraspanin (CD9) and express CD81+, CD90+, and CD144+. The vesicle size ranged between 145 and 162 nm, with the control group exhibiting smaller vesicles (D10) than the AMI group. Furthermore, all vesicles expressed CCR6 and CXCR3, whereas a small percentage expressed CCR4. In addition, a decrease in CXCR3 and CCR6 expression was observed in coronary and peripheral AMI blood vesicles compared with the control; however, no difference was found between AMI coronary and AMI peripheral blood vesicles. In conclusion, our study demonstrates, for the first time, changes in the number of extracellular vesicles expressing CD144+, CXCR3, and CCR6 in the peripheral circulation of patients with AMI. Extracellular vesicles present in the circulation of patients with AMI hold excellent promise as a potential diagnostic tool.
Collapse
Affiliation(s)
- Alexa Moreno
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
- Clinical Laboratory Program, Faculty of Health Sciences, State University of Southern Manabí, Jipijapa 130402, Ecuador
| | - Pedro Alarcón-Zapata
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Enrique Guzmán-Gútierrez
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Claudia Radojkovic
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Héctor Contreras
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Estefanía Nova-Lampeti
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Felipe A Zúñiga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Llerenty Rodriguez-Alvárez
- Department of Animal Science, Faculty of Veterinary Sciences, University of Concepcion, Chillán 3780000, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bio-Bio, Chillán 3780000, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3780000, Chile
| | - Paola Lagos
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
| | - Claudio Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepcion, P.O. Box 237, Concepción 4030000, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán 3780000, Chile
| |
Collapse
|
3
|
Bhat OM, Mir RA, Nehvi IB, Wani NA, Dar AH, Zargar MA. Emerging role of sphingolipids and extracellular vesicles in development and therapeutics of cardiovascular diseases. IJC HEART & VASCULATURE 2024; 53:101469. [PMID: 39139609 PMCID: PMC11320467 DOI: 10.1016/j.ijcha.2024.101469] [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: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Sphingolipids are eighteen carbon alcohol lipids synthesized from non-sphingolipid precursors in the endoplasmic reticulum (ER). The sphingolipids serve as precursors for a vast range of moieties found in our cells that play a critical role in various cellular processes, including cell division, senescence, migration, differentiation, apoptosis, pyroptosis, autophagy, nutrition intake, metabolism, and protein synthesis. In CVDs, different subclasses of sphingolipids and other derived molecules such as sphingomyelin (SM), ceramides (CERs), and sphingosine-1-phosphate (S1P) are directly related to diabetic cardiomyopathy, dilated cardiomyopathy, myocarditis, ischemic heart disease (IHD), hypertension, and atherogenesis. Several genome-wide association studies showed an association between genetic variations in sphingolipid pathway genes and the risk of CVDs. The sphingolipid pathway plays an important role in the biogenesis and secretion of exosomes. Small extracellular vesicles (sEVs)/ exosomes have recently been found as possible indicators for the onset of CVDs, linking various cellular signaling pathways that contribute to the disease progression. Important features of EVs like biocompatibility, and crossing of biological barriers can improve the pharmacokinetics of drugs and will be exploited to develop next-generation drug delivery systems. In this review, we have comprehensively discussed the role of sphingolipids, and sphingolipid metabolites in the development of CVDs. In addition, concise deliberations were laid to discuss the role of sEVs/exosomes in regulating the pathophysiological processes of CVDs and the exosomes as therapeutic targets.
Collapse
Affiliation(s)
- Owais Mohmad Bhat
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | | | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - Abid Hamid Dar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| | - M Afzal Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, India
| |
Collapse
|
4
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
5
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
6
|
McDonald J, Mohak S, Fabian Z. Stem Cell-Derived Extracellular Vesicles in the Treatment of Cardiovascular Diseases. Pharmaceutics 2024; 16:381. [PMID: 38543275 PMCID: PMC10974254 DOI: 10.3390/pharmaceutics16030381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 01/03/2025] Open
Abstract
Cardiovascular disease constitutes a noteworthy public health challenge characterized by a pronounced incidence, frequency, and mortality rate, particularly impacting specific demographic groups, and imposing a substantial burden on the healthcare infrastructure. Certain risk factors, such as age, gender, and smoking, contribute to the prevalence of fatal cardiovascular disease, highlighting the need for targeted interventions. Current challenges in clinical practice involve medication complexities, the lack of a systematic decision-making approach, and prevalent drug therapy problems. Stem cell-derived extracellular vesicles stand as versatile entities with a unique molecular fingerprint, holding significant therapeutic potential across a spectrum of applications, particularly in the realm of cardio-protection. Their lipid, protein, and nucleic acid compositions, coupled with their multifaceted functions, underscore their role as promising mediators in regenerative medicine and pave the way for further exploration of their intricate contributions to cellular physiology and pathology. Here, we overview our current understanding of the possible role of stem cell-derived extracellular vesicles in the clinical management of human cardiovascular pathologies.
Collapse
Affiliation(s)
- Jennifer McDonald
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Fylde Road, Preston PR1 2HE, UK;
| | - Sidhesh Mohak
- Department of Internal Medicine, South Texas Health System, McAllen, TX 78503, USA;
| | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Fylde Road, Preston PR1 2HE, UK;
| |
Collapse
|
7
|
Jiang J, Zhang X, Wang H, Spanos M, Jiang F, Ni L, Li J, Li G, Lin Y, Xiao J. Closer to The Heart: Harnessing the Power of Targeted Extracellular Vesicle Therapies. Adv Biol (Weinh) 2024; 8:e2300141. [PMID: 37953665 DOI: 10.1002/adbi.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 09/08/2023] [Indexed: 11/14/2023]
Abstract
Extracellular vesicles (EVs) have emerged as novel diagnostic and therapeutic approaches for cardiovascular diseases. EVs derived from various origins exhibit distinct effects on the cardiovascular system. However, the application of native EVs is constrained due to their poor stabilities and limited targeting capabilities. Currently, targeted modification of EVs primarily involves genetic engineering, chemical modification (covalent, non-covalent), cell membrane modification, and biomaterial encapsulation. These techniques enhance the stability, biological activity, target-binding capacity, and controlled release of EVs at specific cells and tissues. The diverse origins of cardioprotective EVs are covered, and the applications of cardiac-targeting EV delivery systems in protecting against cardiovascular diseases are discussed. This review summarizes the current stage of research on the potential of EV-based targeted therapies for addressing cardiovascular disorders.
Collapse
Affiliation(s)
- Jizong Jiang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xinxin Zhang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Hongyun Wang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Fei Jiang
- Department of Nursing, Union Hospital, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Lingyan Ni
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jin Li
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yanjuan Lin
- Department of Nursing, Union Hospital, Fujian Medical University Union Hospital, Fuzhou, 350001, China
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
8
|
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
|
9
|
Zheng C, Wu Y, Xu J, Liu Y, Ma J. Exosomes from bone marrow mesenchymal stem cells ameliorate glucocorticoid-induced osteonecrosis of femoral head by transferring microRNA-210 into bone microvascular endothelial cells. J Orthop Surg Res 2023; 18:939. [PMID: 38062514 PMCID: PMC10704824 DOI: 10.1186/s13018-023-04440-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVES Bone microvascular endothelial cells (BMECs) played an important role in the pathogenesis of glucocorticoid-induced osteonecrosis of femoral head (GCS-ONFH), and exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos) may provide an effective treatment. This study aimed to evaluate the effects of BMSC-Exos and internal microRNA-210-3p (miRNA-210) on GCS-ONFH in an in vitro hydrocortisone-induced BMECs injury model and an in vivo rat GCS-ONFH model. METHODS BMECs, BMSCs and BMSC-Exos were isolated and validated. BMECs after the treatment of hydrocortisone were cocultured with different concentrations of BMSC-Exos, then proliferation, migration, apoptosis and angiogenesis of BMECs were evaluated by CCK-8, Annexin V-FITC/PI, cell scratch and tube formation assays. BMSCs were transfected with miRNA-210 mimics and miRNA-210 inhibitors, then BMSC-ExosmiRNA-210 mimic and BMSC-ExosmiRNA-210 inhibitor secreted from such cells were collected. The differences between BMSC-Exos, BMSC-ExosmiRNA-210 mimic and BMSC-ExosmiRNA-210 inhibitor in protecting BMECs against GCS treatment were analyzed by methods mentioned above. Intramuscular injections of methylprednisolone were performed on Sprague-Dawley rats to establish an animal model of GCS-ONFH, then tail intravenous injections of BMSC-Exos, BMSC-ExosmiRNA-210 mimic or BMSC-ExosmiRNA-210 inhibitor were conducted after methylprednisolone injection. Histological and immunofluorescence staining and micro-CT were performed to evaluate the effects of BMSC-Exos and internal miRNA-210 on the in vivo GCS-ONFH model. RESULTS Different concentrations of BMSC-Exos, especially high concentration of BMSC-Exos, could enhance the proliferation, migration and angiogenesis ability and reduce the apoptosis rates of BMECs treated with GCS. Compared with BMSC-Exos, BMSC-ExosmiRNA-210 mimic could further enhance the proliferation, migration and angiogenesis ability and reduce the apoptosis rates of BMECs, while BMECs in the GCS + BMSC-ExosmiRNA-210 inhibitor group showed reduced proliferation, migration and angiogenesis ability and higher apoptosis rates. In the rat GCS-ONFH model, BMSC-Exos, especially BMSC-ExosmiRNA-210 mimic, could increase microvascular density and enhance bone remodeling of femoral heads. CONCLUSIONS BMSC-Exos containing miRNA-210 could serve as potential therapeutics for protecting BMECs and ameliorating the progression of GCS-ONFH.
Collapse
Affiliation(s)
- Che Zheng
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 37# Guoxue Road, Chengdu, 610041, People's Republic of China
- Department of Orthopedic Surgery, Chengdu Second People's Hospital, Chengdu, Sichuan Province, People's Republic of China
| | - Yuangang Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 37# Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Jiawen Xu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 37# Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Yuan Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 37# Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Jun Ma
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, 37# Guoxue Road, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
10
|
Wagh V, Nguemo F, Kiseleva Z, Mader RM, Hescheler J, Mohl W. Circulating microRNAs and cardiomyocyte proliferation in heart failure patients related to 10 years survival. ESC Heart Fail 2023; 10:3559-3572. [PMID: 37752740 PMCID: PMC10682869 DOI: 10.1002/ehf2.14516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/17/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023] Open
Abstract
AIMS Mechanochemical signalling drives organogenesis and is highly conserved in mammal evolution. Regaining recovery in myocardial jeopardy by inducing principles linking cardiovascular therapy and clinical outcome has been the dream of scientists for decades. Concepts involving embryonic pathways to regenerate adult failing hearts became popular in the early millennium. Since then, abundant data on stem cell research have been published, never reaching widespread application in heart failure therapy. Another conceptual access, using mechanotransduction in cardiac veins to limit myocardial decay, is pressure-controlled intermittent coronary sinus occlusion (PICSO). Recently, we reported acute molecular signs and signals of PICSO activating regulatory miRNA and inducing cell proliferation mimicking cardiac development in adult failing hearts. According to a previously formulated hypothesis, 'embryonic recall', this study aimed to define molecular signals involved in endogenous heart repair during PICSO and study their relation to patient survival. METHODS AND RESULTS We previously reported a study on the acute molecular effects of PICSO in an observational non-randomized study. Eight out of the thirty-two patients with advanced heart failure undergoing cardiac resynchronization therapy (CRT) were treated with PICSO. Survival was monitored over 10 years, and coronary sinus blood samples were collected during intervention before and after 20 min and tested for miRNA signalling and proliferation when co-cultured with cardiomyocytes. A numerically lower death rate post-CRT and PICSO as compared with control CRT only, and a non-significant reduction in all-cause mortality risk of 42% was observed (37.5% vs. 54.0%, relative risk = 0.58, 95% confidence interval: 0.17-2.05; P = 0.402). Four miRNAs involved in cell cycle, proliferation, morphogenesis, embryonic development, and apoptosis significantly increased concomitantly in survivors and PICSO compared with a decrease in non-survivors (hsa-miR Let7b, P < 0.01; hsa-miR- 421, P < 0.006; hsa-miR 363-3p, P < 0.03 and hsa-miR 19b-3p P < 0.01). In contrast, three miRNAs involved in proliferation and survival, determining cell fate, and recycling endosomes decreased in survivors and PICSO (hsa miR 101-3p, P < 0.03; hsa-miR 25-3p, P < 002; hsa-miR 30d-5p P < 0.04). In vitro cellular proliferation increased in survivors and lowered in non-survivors showing a pattern distinction, discriminating longevity according to up to 10-year survival in heart failure patients. CONCLUSIONS This study proposes that generating regenerative signals observed during PICSO intervention relate to patient outcomes. Morphogenetic pathways induced by periods of flow reversal in cardiac veins in a domino-like pattern transform embryonic into regenerative signals. Studies supporting the conversion of mechanochemical signals into regenerative molecules during PICSO are warranted to substantiate predictive power on patient longevity, opening new therapeutic avenues in otherwise untreatable heart failure.
Collapse
Affiliation(s)
- Vilas Wagh
- Merck Research LabsBostonMAUSA
- Center of Physiology and Pathophysiology, Institute of NeurophysiologyUniversity of CologneCologneGermany
| | - Filomain Nguemo
- Center of Physiology and Pathophysiology, Institute of NeurophysiologyUniversity of CologneCologneGermany
| | - Zlata Kiseleva
- Department of Cardiac Surgery emeritusMedical University ViennaViennaAustria
| | - Robert M. Mader
- Department of Medicine IMedical University ViennaViennaAustria
| | - Juergen Hescheler
- Center of Physiology and Pathophysiology, Institute of NeurophysiologyUniversity of CologneCologneGermany
| | - Werner Mohl
- Department of Cardiac Surgery emeritusMedical University ViennaViennaAustria
| |
Collapse
|
11
|
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
|
12
|
Li J, Song F, Chen R, Yang J, Liu J, Huang L, Duan F, Kou M, Lian BX, Zhou X, Han W, Mao L, Wu C, Wu W, Wei R, Chen H, Xu A, Tse HF, Lian Q, Li G, Wang Y. Bradykinin-pretreated Human cardiac-specific c-kit + Cells Enhance Exosomal miR-3059-5p and Promote Angiogenesis Against Hindlimb Ischemia in mice. Stem Cell Rev Rep 2023; 19:2481-2496. [PMID: 37535186 DOI: 10.1007/s12015-023-10591-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Protection of cardiac function following myocardial infarction was largely enhanced by bradykinin-pretreated cardiac-specific c-kit+ (BK-c-kit+) cells, even without significant engraftment, indicating that paracrine actions of BK-c-kit+ cells play a pivotal role in angiogenesis. Nevertheless, the active components of the paracrine actions of BK-c-kit+ cells and the underlying mechanisms remain unknown. This study aimed to define the active components of exosomes from BK-c-kit+ cells and elucidate their underlying protective mechanisms. METHODS Matrigel tube formation assay, cell cycle, and mobility in human umbilical vein endothelial cells (HUVECs) and hindlimb ischemia (HLI) in mice were applied to determine the angiogenic effect of condition medium (CM) and exosomes. Proteome profiler, microRNA sponge, Due-luciferase assay, microRNA-sequencing, qRT-PCR, and Western blot were used to determine the underlying mechanism of the angiogenic effect of exosomes from BK-c-kit+. RESULTS As a result, BK-c-kit+ CM and exosomes promoted tube formation in HUVECs and the repair of HLI in mice. Angiogenesis-related proteomic profiling and microRNA sequencing revealed highly enriched miR-3059-5p as a key angiogenic component of BK-c-kit+ exosomes. Meanwhile, loss- and gain-of-function experiments revealed that the promotion of angiogenesis by miR-3059-5p was mainly through suppression of TNFSF15-inhibited effects on vascular tube formation, cell proliferation and cell migration. Moreover, enhanced angiogenesis of miR-3059-5p-inhibited TNFSF15 has been associated with Akt/Erk1/2/Smad2/3-modulated signaling pathway. CONCLUSION Our results demonstrated a novel finding that BK-c-kit+ cells enrich exosomal miR-3059-5p to suppress TNFSF15 and promote angiogenesis against hindlimb ischemia in mice.
Collapse
Affiliation(s)
- Jingzhou Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Fei Song
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ruolan Chen
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinjuan Yang
- Cord Blood Bank, Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jie Liu
- Cord Blood Bank, Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Medicine, State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Li Huang
- Cord Blood Bank, Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fuyu Duan
- Cord Blood Bank, Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Meng Kou
- Cord Blood Bank, Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Boon Xuan Lian
- University of Adelaide Medical School, Adelaide, Australia
| | - Xiaoxia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Weimin Han
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Liang Mao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Weiyin Wu
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Rui Wei
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China
| | - Aimin Xu
- Department of Medicine, State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- Department of Medicine, State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
| | - Qizhou Lian
- Cord Blood Bank, Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Medicine, State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China.
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, State Key Laboratory of Pharmaceutical Biotechnology, Chinese Academy of Sciences, the University of Hong Kong, Shenzhen, China.
| | - Gang Li
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
13
|
Kore RA, Jenkins SV, Jamshidi-Parsian A, Tackett AJ, Griffin RJ, Ayyadevara S, Mehta JL. Proteomic analysis of transcription factors involved in the alteration of ischemic mouse heart as modulated by MSC exosomes. Biochem Biophys Rep 2023; 34:101463. [PMID: 37125076 PMCID: PMC10130341 DOI: 10.1016/j.bbrep.2023.101463] [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: 02/07/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Mesenchymal stem cell (MSC) exosomes have been found to attenuate cardiac systolic and diastolic dysfunction in animal models of ischemia. Exosomes carry a plethora of active and inactive proteins as their cargo, which are readily available to the recipient cell for use in intracellular signaling pathways-depending on the stresses, such as ischemia or hypoxia. Among the exosomal proteins are the often-overlooked cargo of transcriptional regulators. These transcriptional regulators influence the transcriptome and subsequently the proteome of recipient cell. Here, we report the transcriptional factors and regulators differentially modulated and their potential role in modulating cardiac function in MSC exosome treated ischemic mice hearts. Our analysis shows ischemic stress modulating transcriptional regulators and factors such as HSF1 and HIF1A in the infarct and peri-infarct areas of ischemic hearts which is mitigated by MSC exosomes. Similarly, STAT3 and SMAD3 are also modulated by MSC exosomes. Interestingly, NOTCH1 and β-catenin were detected in the ischemic hearts. The differential expression of these regulators and factors drives changes in various biological process governed in the ischemic cardiac cells. We believe these studies will advance our understanding of cardiac dysfunction occurring in the ischemic hearts and lay the groundwork for further studies on the modulation of cardiac function during ischemia by MSC exosomes.
Collapse
Affiliation(s)
- Rajshekhar A. Kore
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Corresponding author.
| | - Samir V. Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Azemat Jamshidi-Parsian
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Alan J. Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Robert J. Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Srinivas Ayyadevara
- Department of Geriatrics, Central Arkansas Veterans Healthcare System and University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Jawahar L. Mehta
- Division of Cardiology, Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
- Corresponding author.
| |
Collapse
|
14
|
Gao C, Xu YJ, Meng ZX, Gu S, Zhang L, Zheng L. BMSC-Derived Exosomes Carrying lncRNA-ZFAS1 Alleviate Pulmonary Ischemia/Reperfusion Injury by UPF1-Mediated mRNA Decay of FOXD1. Mol Neurobiol 2023; 60:2379-2396. [PMID: 36652050 DOI: 10.1007/s12035-022-03129-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/04/2022] [Indexed: 01/19/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) exert protective effects against pulmonary ischemia/reperfusion (I/R) injury; however, the potential mechanism involved in their protective ability remains unclear. Thus, this study aimed to explore the function and underlying mechanism of BMSC-derived exosomal lncRNA-ZFAS1 in pulmonary I/R injury. Pulmonary I/R injury models were established in mice and hypoxia/reoxygenation (H/R)-exposed primary mouse lung microvascular endothelial cells (LMECs). Exosomes were extracted from BMSCs. Target molecule expression was assessed by qRT-PCR and Western blotting. Pathological changes in the lungs, pulmonary edema, apoptosis, pro-inflammatory cytokine levels, SOD, MPO activities, and MDA level were measured. The proliferation, apoptosis, and migration of LMECs were detected by CCK-8, EdU staining, flow cytometry, and scratch assay. Dual-luciferase reporter assay, RNA pull-down, RIP, and ChIP assays were performed to validate the molecular interaction. In the mouse model of pulmonary I/R injury, BMSC-Exos treatment relieved lung pathological injury, reduced lung W/D weight ratio, and restrained apoptosis and inflammation, whereas exosomal ZFAS1 silencing abolished these beneficial effects. In addition, the proliferation, migration inhibition, apoptosis, and inflammation in H/R-exposed LMECs were repressed by BMSC-derived exosomal ZFAS1. Mechanistically, ZFAS1 contributed to FOXD1 mRNA decay via interaction with UPF1, thereby leading to Gal-3 inactivation. Furthermore, FOXD1 depletion strengthened the weakened protective effect of ZFAS1-silenced BMSC-Exos on pulmonary I/R injury. ZFAS1 delivered by BMSC-Exos results in FOXD1 mRNA decay and subsequent Gal-3 inactivation via direct interaction with UPF1, thereby attenuating pulmonary I/R injury.
Collapse
Affiliation(s)
- Cao Gao
- Departments of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Yan-Jie Xu
- Departments of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Zhi-Xiu Meng
- Departments of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Shuang Gu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Road, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Lei Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Road, Changzhou, 213000, Jiangsu Province, People's Republic of China
| | - Liang Zheng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, No. 185, Juqian Road, Changzhou, 213000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
15
|
Effects of Human Deciduous Dental Pulp-Derived Mesenchymal Stem Cell-Derived Conditioned Medium on the Metabolism of HUVECs, Osteoblasts, and BMSCs. Cells 2022; 11:cells11203222. [PMID: 36291089 PMCID: PMC9600042 DOI: 10.3390/cells11203222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we assessed the effects of human deciduous dental pulp-derived mesenchymal stem cell-derived conditioned medium (SHED-CM) on the properties of various cell types. The effects of vascular endothelial growth factor (VEGF) in SHED-CM on the luminal architecture, proliferative ability, and angiogenic potential of human umbilical vein endothelial cells (HUVECs) were determined. We also investigated the effects of SHED-CM on the proliferation of human-bone-marrow mesenchymal stem cells (hBMSCs) and mouse calvarial osteoblastic cells (MC3T3-E1) as well as the expression of ALP, OCN, and RUNX2. The protein levels of ALP were examined using Western blot analysis. VEGF blockade in SHED-CM suppressed the proliferative ability and angiogenic potential of HUVECs, indicating that VEGF in SHED-CM contributes to angiogenesis. The culturing of hBMSCs and MC3T3-E1 cells with SHED-CM accelerated cell growth and enhanced mRNA expression of bone differentiation markers. The addition of SHED-CM enhanced ALP protein expression in hBMSCs and MT3T3-E1 cells compared with that of the 0% FBS group. Furthermore, SHED-CM promoted the metabolism of HUVECs, MC3T3-E1 cells, and hBMSCs. These findings indicate the potential benefits of SHED-CM in bone tissue regeneration.
Collapse
|
16
|
Xu J, Wang W, Wang Y, Zhu Z, Li D, Wang T, Liu K. Progress in research on the role of exosomal miRNAs in the diagnosis and treatment of cardiovascular diseases. Front Genet 2022; 13:929231. [PMID: 36267409 PMCID: PMC9577319 DOI: 10.3389/fgene.2022.929231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022] Open
Abstract
Cardiovascular diseases are the most common diseases threatening the health of the elderly, and the incidence and mortality rates associated with cardiovascular diseases remain high and are increasing gradually. Studies on the treatment and prevention of cardiovascular diseases are underway. Currently, several research groups are studying the role of exosomes and biomolecules incorporated by exosomes in the prevention, diagnosis, and treatment of clinical diseases, including cardiovascular diseases. Now, based on the results of published studies, this review discusses the characteristics, separation, extraction, and identification of exosomes, specifically the role of exosomal miRNAs in atherosclerosis, myocardial injury and infarction, heart failure, aortic dissection, myocardial fibrosis, ischemic reperfusion, atrial fibrillation, and other diseases. We believe that the observations noted in this article will aid in the prevention, diagnosis, and treatment of cardiovascular diseases.
Collapse
|
17
|
Ma J, Shen M, Yue D, Wang W, Gao F, Wang B. Extracellular Vesicles from BMSCs Prevent Glucocorticoid-Induced BMECs Injury by Regulating Autophagy via the PI3K/Akt/mTOR Pathway. Cells 2022; 11:2104. [PMID: 35805188 PMCID: PMC9265732 DOI: 10.3390/cells11132104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 12/17/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a common clinical disease with a high disability rate. Injury of bone microvascular endothelial cells (BMECs) caused by glucocorticoid administration is one of the important causes of ONFH, and there is currently a lack of effective clinical treatments. Extracellular vesicles derived from bone stem cells (BMSC-EVs) can prevent ONFH by promoting angiogenesis and can inhibit cell apoptosis by regulating autophagy via the PI3K/Akt/mTOR signaling pathway. The present study aimed to investigate the effect of extracellular vesicles derived from bone marrow stem cells (BMSC) on a glucocorticoid-induced injury of BMECs and possible mechanisms. We found that BMSC-EVs attenuated glucocorticoid-induced viability, angiogenesis capacity injury, and the apoptosis of BMECs. BMSC-EVs increased the LC3 level, but decreased p62 (an autophagy protein receptor) expression, suggesting that BMSC-Exos activated autophagy in glucocorticoid-treated BMECs. The protective effects of BMSC-EVs on the glucocorticoid-induced injury of BMECs was mimicked by a known stimulator of autophagy (rapamycin) and could be enhanced by co-treatment with an autophagy inhibitor (LY294002). BMSC-EVs also suppressed the PI3K/Akt/mTOR signaling pathway, which regulates cell autophagy, in glucocorticoid-treated BMECs. In conclusion, the results indicate that BMSC-EVs prevent the glucocorticoid-induced injury of BMECs by regulating autophagy via the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Jinhui Ma
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Mengran Shen
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China;
| | - Debo Yue
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Weiguo Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Fuqiang Gao
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| | - Bailiang Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing 100029, China; (J.M.); (D.Y.); (W.W.)
| |
Collapse
|
18
|
Tang J, Cui X, Zhang Z, Xu Y, Guo J, Soliman BG, Lu Y, Qin Z, Wang Q, Zhang H, Lim KS, Woodfield TBF, Zhang J. Injection-Free Delivery of MSC-Derived Extracellular Vesicles for Myocardial Infarction Therapeutics. Adv Healthc Mater 2022; 11:e2100312. [PMID: 34310068 DOI: 10.1002/adhm.202100312] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/09/2021] [Indexed: 12/17/2022]
Abstract
As emerging therapeutic factors, extracellular vesicles (EVs) offer significant potential for myocardial infarction (MI) treatment. Current delivery approaches for EVs involve either intra-myocardial or intravenous injection, where both have inherent limitations for downstream clinical applications such as secondary tissue injury and low delivery efficiency. Herein, an injection-free approach for delivering EVs onto the heart surface to treat MI is proposed. By spraying a mixture of EVs, gelatin methacryloyl (GelMA) precursors, and photoinitiators followed by visible light irradiation for 30 s, EVs are physically entrapped within the GelMA hydrogel network covering the surface of the heart, resulting in an enhanced retention rate. Moreover, EVs are gradually released from the hydrogel network through a combination of diffusion and/or enzymatic degradation of the hydrogel, and they are effectively taken up by the sprayed tissue area. More importantly, the released EVs further migrate deep into myocardium tissue, which exerts an improved therapeutic effect. In an MI-induced mice model, the group treated with EVs-laden GelMA hydrogels shows significant recovery in cardiac function after 4 weeks. The work demonstrates a new strategy for delivering EVs into cardiac tissues for MI treatment in a localized manner with high retention.
Collapse
Affiliation(s)
- Junnan Tang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Zenglei Zhang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Yanyan Xu
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Jiacheng Guo
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Bram G Soliman
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Yongzheng Lu
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Zhen Qin
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials Sichuan University Chengdu Sichuan 61004 China
| | - Hu Zhang
- Henry E. Riggs School of Applied Life Sciences Keck Graduate Institute Claremont CA 91711 USA
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group Department of Orthopaedic Surgery & Musculoskeletal Medicine University of Otago Christchurch 8011 New Zealand
| | - Jinying Zhang
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
- Henan Province Key Laboratory of Cardiac Injury and Repair Zhengzhou Henan 450052 China
| |
Collapse
|
19
|
|
20
|
Kang H, Bae Y, Kwon Y, Kim S, Park J. Extracellular Vesicles Generated Using Bioreactors and their Therapeutic Effect on the Acute Kidney Injury Model. Adv Healthc Mater 2022; 11:e2101606. [PMID: 34773445 DOI: 10.1002/adhm.202101606] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/09/2021] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) are nano-sized vesicles secreted by cells, having beneficial effects for various types of regenerative processes. Although EVs have shown promising effects as therapeutic agents, these effects are difficult to research due to the limitations of EV production. In this study, an EV production method based on a flat-plate bioreactor is introduced. The bioreactor produces approximately seven times more mesenchymal stem cell-derived EVs than static culture conditions. The mechanism underlying the increased production of EVs in a flat-plate bioreactor and its application to acute kidney injury is investigated. This study describes the mechanism of EV production by demonstrating the link between EV biogenesis and increased calcium ion concentration under flow conditions. EVs secreted by cells cultured in the bioreactor have therapeutic efficacy in terms of improving kidney damage, resulting in tissue regeneration in a cisplatin-induced acute kidney injury model. This method will help overcome the limitations of EV production, and the analysis of the application of EVs will increase their reliability as well as the understanding of the use of bioreactor-derived EVs as therapeutic agents.
Collapse
Affiliation(s)
- Hyejin Kang
- School of Interdisciplinary Bioscience and Bioengineering Pohang University of Science and Technology Pohang Gyeongbuk 37673 Republic of Korea
| | - Yeon‐hee Bae
- EXOSOMEplus Inc. Suwon Gyeonggi‐do 16229 Republic of Korea
| | - Yongmin Kwon
- Department of Mechanical Engineering Pohang University of Science and Technology Pohang Gyeongbuk 37673 Republic of Korea
| | - Sejoong Kim
- Department of Internal Medicine Seoul National University Bundang Hospital Pohang Gyeonggi‐do 13620 Republic of Korea
| | - Jaesung Park
- School of Interdisciplinary Bioscience and Bioengineering Pohang University of Science and Technology Pohang Gyeongbuk 37673 Republic of Korea
- Department of Mechanical Engineering Pohang University of Science and Technology Pohang Gyeongbuk 37673 Republic of Korea
| |
Collapse
|
21
|
Cardiac microvascular functions improved by MSC-derived exosomes attenuate cardiac fibrosis after ischemia-reperfusion via PDGFR-β modulation. Int J Cardiol 2021; 344:13-24. [PMID: 34534604 DOI: 10.1016/j.ijcard.2021.09.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/17/2021] [Accepted: 09/09/2021] [Indexed: 01/04/2023]
Abstract
Microvascular dysfunction caused by cardiac ischemia-reperfusion (I/R) leads to multiple severe cardiac adverse events, such as heart failure and ventricular modeling, which plays a critical role in outcomes. Though marrow mesenchymal stem cell (MSC) therapy has been proven effective for attenuating I/R injury, the limitations of clinical feasibility cannot be ignored. Since exosomes are recognized as the main vehicles for MSCs paracrine effects, we assumed that MSC-derived exosomes could prevent microvascular dysfunction and further protect cardiac function. By establishing a rat cardiac I/R model in vivo and a cardiac microvascular endothelial cells (CMECs) hypoxia-reperfusion (H/R) model in vitro, we demonstrated that MSC-derived exosomes enhanced microvascular regeneration under stress, inhibited fibrosis development, and eventually improved cardiac function through platelet-derived growth factor receptor-β (PDGFR-β) modulation. Furthermore, we found that MSC-derived exosomes possessed better therapeutic effects than MSCs themselves.
Collapse
|
22
|
Ma W, Zhang X, Liu Y. miR-124 promotes apoptosis and inhibits the proliferation of vessel endothelial cells through P38/MAPK and PI3K/AKT pathways, making it a potential mechanism of vessel endothelial injury in acute myocardial infarction. Exp Ther Med 2021; 22:1383. [PMID: 34650631 PMCID: PMC8506947 DOI: 10.3892/etm.2021.10819] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/11/2021] [Indexed: 11/26/2022] Open
Abstract
Due to its rapid onset and high rates of fatality, acute myocardial infarction (AMI) has long been one of the most fatal diseases among all types of heart diseases. Therefore, intensive research efforts have been focused on understanding AMI's potential pathogenesis to seek effective treatment options. In the present study, 20 peripheral blood samples were collected from patients with AMI, after which reverse transcription-quantitative PCR analysis revealed that microRNA (miR)-124 levels in the peripheral blood of patients with AMI was significantly elevated compared with that in the control group. In vitro, a model using pcDNA3.1-miR-124 transfected human umbilical vein endothelial cells (HUVECs) indicated that overexpression of miR-124 could significantly promote the apoptosis and suppress the proliferation of HUVECs using flow cytometry, TUNEL assay and Cell Counting Kit-8 assays. Based on the present findings, RNA samples of HUVECs overexpressing miR-124 was extracted and sequenced to explore the gene expression profile after miR-124 overexpression. Gene Set Enrichment Analysis (GSEA) analysis revealed that the downregulated genes were mainly enriched in signaling pathways, such as PI3K-AKT, whilst the upregulated genes were mainly enriched in metabolism-related signaling pathways, such as the metabolism of xenobiotics by cytochrome P450 pathway. Additionally, Rideogram software was used to determine the chromosomal localization of the differentially expressed genes. The results demonstrated that they were distributed on all chromosomes except for chromosome Y. In addition, characteristic profiles of the differentially expressed genes caused by miR-124 overexpression were analyzed using Connectivity Map. In total, two medicines, anisomycin and sanguinarine, which function as p38/MAPK signaling agonists that can inhibit angiogenesis, presented with the highest enrichment scores. Together with the GSEA results, which indicated that the differentially expressed genes were mainly enriched in the angiogenesis-inhibiting PI3K/AKT signaling pathway, the present study reported that high expression of miR-124 was negatively associated with patients with AMI, promoting the apoptosis and suppressing the proliferation of vessel endothelial cells.
Collapse
Affiliation(s)
- Weimin Ma
- Department of Critical Care Medicine, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Xin Zhang
- Department of Pediatrics, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Yang Liu
- Department of Critical Care Medicine, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| |
Collapse
|
23
|
MicroRNAs-The Heart of Post-Myocardial Infarction Remodeling. Diagnostics (Basel) 2021; 11:diagnostics11091675. [PMID: 34574016 PMCID: PMC8469128 DOI: 10.3390/diagnostics11091675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is one of the most frequent cardiac emergencies, with significant potential for mortality. One of the major challenges of the post-MI healing response is that replacement fibrosis could lead to left ventricular remodeling (LVR) and heart failure (HF). This process involves canonical and non-canonical transforming growth factor-beta (TGF-β) signaling pathways translating into an intricate activation of cardiac fibroblasts and disproportionate collagen synthesis. Accumulating evidence has indicated that microRNAs (miRNAs) significantly contribute to the modulation of these signaling pathways. This review summarizes the recent updates regarding the molecular mechanisms underlying the role of the over 30 miRNAs involved in post-MI LVR. In addition, we compare the contradictory roles of several multifunctional miRNAs and highlight their potential use in pressure overload and ischemia-induced fibrosis. Finally, we discuss their attractive role as prognostic biomarkers for HF, highlighting the most relevant human trials involving these miRNAs.
Collapse
|
24
|
Yao J, Huang K, Zhu D, Chen T, Jiang Y, Zhang J, Mi L, Xuan H, Hu S, Li J, Zhou Y, Cheng K. A Minimally Invasive Exosome Spray Repairs Heart after Myocardial Infarction. ACS NANO 2021; 15:11099-11111. [PMID: 34152126 DOI: 10.1021/acsnano.1c00628] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Myocardial infarction (MI) remains the most common cause of death worldwide. Many MI survivors will suffer from recurrent heart failure (HF), which has been recognized as a determinant of adverse prognosis. Despite the success of improved early survival after MI by primary percutaneous coronary intervention, HF after MI is becoming the major driver of late morbidity, mortality, and healthcare costs. The development of regenerative medicine has brought hope to MI treatment in the past decade. Mesenchymal stem cell (MSC)-derived exosomes have been established as an essential part of stem cell paracrine factors for heart regeneration. However, its regenerative power is hampered by low delivery efficiency to the heart. We designed, fabricated, and tested a minimally invasive exosome spray (EXOS) based on MSC exosomes and biomaterials. In a mouse model of acute myocardial infarction, EXOS improved cardiac function and reduced fibrosis, and promoted endogenous angiomyogenesis in the post-injury heart. We further tested the feasibility and safety of EXOS in a pig model. Our results indicate that EXOS is a promising strategy to deliver therapeutic exosomes for heart repair.
Collapse
Affiliation(s)
- Jialu Yao
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Tan Chen
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Junyi Zhang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Lijie Mi
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - He Xuan
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Junlang Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Yafeng Zhou
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
25
|
Abstract
The role of stem cells in augmenting reparative processes in the heart after ischemic injury has been successfully demonstrated in small and large animal models. However, the outcomes of cell therapy in clinical trials have been somewhat variable, with overall effects of autologous stem cell therapies demonstrating a modest improvement in cardiac structure and function. How stem cells repair the heart after cardiac injury is still not well understood. Most recent studies suggest that adult derived stem cells act primarily through paracrine signaling to exert beneficial effects, including modulation of immune response, stimulation of new blood vessel formation, or by inducing mature myocytes to transiently reenter the cell cycle, rather than robust direct differentiation of the transplanted cells into myocytes. In addition, data from multiple laboratory results confirmed clearance of stem cells themselves within a few days still leading to functional benefits further confirming the role of paracrine signaling in augmenting cardiac reparative processes rather than direct differentiation of cells. These findings rapidly evolved the field of extracellular vesicles specifically microvesicles (MVs) as they are active hubs of autocrine, paracrine, and endocrine signaling targeting different biological processes. The beneficial effects seen after stem cell transplantation could be linked to the cardioprotective factors packaged in the MVs secreted from stem cells. Therefore, stem cell MVs provide a new avenue for the treatment of cardiovascular disease through a multitude of mechanisms including cellular communication within the stem cell niches, delivery of genetic information, regulation of the immune system in the heart, and stimulation of angiogenesis which will be discussed in this review.
Collapse
|
26
|
Ghodrat S, Hoseini SJ, Asadpour S, Nazarnezhad S, Alizadeh Eghtedar F, Kargozar S. Stem cell-based therapies for cardiac diseases: The critical role of angiogenic exosomes. Biofactors 2021; 47:270-291. [PMID: 33606893 DOI: 10.1002/biof.1717] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
Finding effective treatments for cardiac diseases is among the hottest subjects in medicine; cell-based therapies have brought great promises for managing a broad range of life-threatening heart complications such as myocardial infarction. After clarifying the critical role of angiogenesis in tissue repair and regeneration, various stem/progenitor cell were utilized to accelerate the healing of injured cardiac tissue. Embryonic, fetal, adult, and induced pluripotent stem cells have shown the appropriate proangiogenic potential for tissue repair strategies. The capability of stem cells for differentiating into endothelial lineages was initially introduced as the primary mechanism involved in improving angiogenesis and accelerated heart tissue repair. However, recent studies have demonstrated the leading role of paracrine factors secreted by stem cells in advancing neo-vessel formation. Genetically modified stem cells are also being applied for promoting angiogenesis regarding their ability to considerably overexpress and secrete angiogenic bioactive molecules. Yet, conducting further research seems necessary to precisely identify molecular mechanisms behind the proangiogenic potential of stem cells, including the signaling pathways and regulatory molecules such as microRNAs. In conclusion, stem cells' pivotal roles in promoting angiogenesis and consequent improved cardiac healing and remodeling processes should not be ignored, especially in the case of stem cell-derived extracellular vesicles.
Collapse
Affiliation(s)
- Sara Ghodrat
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Javad Hoseini
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shiva Asadpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Alizadeh Eghtedar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Rheault-Henry M, White I, Grover D, Atoui R. Stem cell therapy for heart failure: Medical breakthrough, or dead end? World J Stem Cells 2021; 13:236-259. [PMID: 33959217 PMCID: PMC8080540 DOI: 10.4252/wjsc.v13.i4.236] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/22/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure continues to be one of the leading causes of morbidity and mortality worldwide. Myocardial infarction is the primary causative agent of chronic heart failure resulting in cardiomyocyte necrosis and the subsequent formation of fibrotic scar tissue. Current pharmacological and non-pharmacological therapies focus on managing symptoms of heart failure yet remain unable to reverse the underlying pathology. Heart transplantation usually cannot be relied on, as there is a major discrepancy between the availability of donors and recipients. As a result, heart failure carries a poor prognosis and high mortality rate. As the heart lacks significant endogenous regeneration potential, novel therapeutic approaches have incorporated the use of stem cells as a vehicle to treat heart failure as they possess the ability to self-renew and differentiate into multiple cell lineages and tissues. This review will discuss past, present, and future clinical trials, factors that influence stem cell therapy outcomes as well as ethical and safety considerations. Preclinical and clinical studies have shown a wide spectrum of outcomes when applying stem cells to improve cardiac function. This may reflect the infancy of clinical trials and the limited knowledge on the optimal cell type, dosing, route of administration, patient parameters and other important variables that contribute to successful stem cell therapy. Nonetheless, the field of stem cell therapeutics continues to advance at an unprecedented pace. We remain cautiously optimistic that stem cells will play a role in heart failure management in years to come.
Collapse
Affiliation(s)
| | - Ian White
- Northern Ontario School of Medicine, Sudbury P3E 2C6, Ontario, Canada
| | - Diya Grover
- Ross University School of Medicine, St. Michael BB11093, Barbados
| | - Rony Atoui
- Division of Cardiac Surgery, Health Sciences North, Northern Ontario School of Medicine, Sudbury P3E 3Y9, Ontario, Canada
| |
Collapse
|
28
|
Agarwal T, Fortunato GM, Hann SY, Ayan B, Vajanthri KY, Presutti D, Cui H, Chan AHP, Costantini M, Onesto V, Di Natale C, Huang NF, Makvandi P, Shabani M, Maiti TK, Zhang LG, De Maria C. Recent advances in bioprinting technologies for engineering cardiac tissue. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112057. [PMID: 33947551 DOI: 10.1016/j.msec.2021.112057] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/12/2022]
Abstract
Annually increasing incidence of cardiac-related disorders and cardiac tissue's minimal regenerative capacity have motivated the researchers to explore effective therapeutic strategies. In the recent years, bioprinting technologies have witnessed a great wave of enthusiasm and have undergone steady advancements over a short period, opening the possibilities for recreating engineered functional cardiac tissue models for regenerative and diagnostic applications. With this perspective, the current review delineates recent developments in the sphere of engineered cardiac tissue fabrication, using traditional and advanced bioprinting strategies. The review also highlights different printing ink formulations, available cellular opportunities, and aspects of personalized medicines in the context of cardiac tissue engineering and bioprinting. On a concluding note, current challenges and prospects for further advancements are also discussed.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Gabriele Maria Fortunato
- Research Center "E. Piaggio" and Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Sung Yun Hann
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Bugra Ayan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Kiran Yellappa Vajanthri
- School of Biomedical Engineering, Indian Institute of Technology Banaras Hindu University Varanasi, Uttar Pradesh 221005, India
| | - Dario Presutti
- Institute of Physical Chemistry - Polish Academy of Sciences, Warsaw, Poland
| | - Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Alex H P Chan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Marco Costantini
- Institute of Physical Chemistry - Polish Academy of Sciences, Warsaw, Poland
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Campus Ecotekne, via Monteroni, Lecce 73100, Italy
| | - Concetta Di Natale
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Naples, Italy; Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.leTecchio 80, Naples 80125, Italy
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Pooyan Makvandi
- Center for Materials Interface, Istituto Italiano di Tecnologia, Pontedera 56025, Pisa, Italy
| | - Majid Shabani
- Center for Materials Interface, Istituto Italiano di Tecnologia, Pontedera 56025, Pisa, Italy
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA; Department of Electrical and Computer Engineering, The George Washington University, Washington, DC 20052, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, USA; Department of Medicine, The George Washington University, Washington, DC 20052, USA.
| | - Carmelo De Maria
- Research Center "E. Piaggio" and Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy.
| |
Collapse
|
29
|
Wang W, Zheng H. Myocardial Infarction: The Protective Role of MiRNAs in Myocardium Pathology. Front Cardiovasc Med 2021; 8:631817. [PMID: 33748196 PMCID: PMC7973051 DOI: 10.3389/fcvm.2021.631817] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases have been regarded as the leading cause of death around the world, with myocardial infarction (MI) being the most severe form. MI leads to myocardial apoptosis, cardiomyocyte fibrosis, and cardiomyocyte hypertrophy, ultimately leading to heart failure, and death. Micro RNAs (miRNAs) participate in the genesis and progression of myocardial pathology after MI by playing an important regulatory role. This review aims to summarize all available knowledge on the role of miRNAs in the myocardial pathological process after MI to uncover potential major target pathways. In addition, the main therapeutic methods and their latest progress are also reviewed. miRNAs can regulate the main signaling pathways as well as pathological processes. Thus, they have the potential to induce therapeutic effects. Hence, the combination of miRNAs with recently developed exosome nanocomplexes may represent the future direction of therapeutics.
Collapse
Affiliation(s)
- Wei Wang
- Graduate School of Bengbu Medical College, Bengbu, China
| | - Hao Zheng
- Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
30
|
MSC exosome-mediated cardioprotection in ischemic mouse heart comparative proteomics of infarct and peri-infarct areas. Mol Cell Biochem 2021; 476:1691-1704. [PMID: 33423165 DOI: 10.1007/s11010-020-04029-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
Mesenchymal stem cell (MSC) exosomes may limit cardiac injury, and even reverse cardiac damage in animal models of ischemia. To understand exosome-mediated improvement in cardiac function we examined the proteomic alternations in the MSC exosome-treated mice hearts subjected to left coronary artery (LCA) ligation, with particular emphasis on peri-infarct areas. At 7 days after LCA ligation, left ventricular end systolic thickness, infarct size and survival of mice were studied. Mass spectrometric analysis of infarct and peri-infarct areas was carried out. Expression of inflammatory markers (LOX-1 and NLRP3) and cell death markers (Bax, Bcl-2, Caspases 1 and 3 and GSDMD) were investigated by Western blots and immunofluorescence. Proteomic analysis of the infarct and peri-infarct areas in saline-treated hearts revealed differentially expressed proteins involved in inflammation and apoptotic cell death, while showing depletion of processes governing cell death. Exosome treatment significantly improved the proteomic profile in both infarct and peri-infarct areas, more so in the peri-infarct areas. The infarct size was smaller (9 ± 1%), and cardiac contractile function (fractional shortening) was preserved in the exosome-treated mice (28 ± 2%). Survival of exosome-treated mice was also better. White blood cell accumulation in and around the infarct area, expression of LOX-1 and NLRP3 inflammasome, and markers of cell death (cleaved Caspase-3, Caspase-1, GSDMD, Bcl-2 and Bax) were dramatically reduced by MSC exosome treatment (all p < 0.01). In cultured primary mouse cardiomyocytes, treatment with MSC exosomes essentially reversed inflammation-induced pro-apoptotic and inflammatory signals (p < 0.01). MSC exosomes exert their cardioprotective effects by suppressing inflammation and pro-apoptotic processes, particularly in the peri-infarct areas, resulting in preservation of cardiac function after LCA ligation.
Collapse
|
31
|
Ye Y, Li SL, Wang JJ. miR-100-5p Downregulates mTOR to Suppress the Proliferation, Migration, and Invasion of Prostate Cancer Cells. Front Oncol 2020; 10:578948. [PMID: 33335853 PMCID: PMC7736635 DOI: 10.3389/fonc.2020.578948] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
Background Previous studies have shown that miR-100-5p expression is abnormal in prostate cancer. However, the role and regulatory mechanism of miR-100-5p requires further investigation. Thus, the aim of this study was to observe the effects of miR-100-5p on the proliferation, migration and invasion of prostate cancer (PCa) cells and to explore the potential related regulatory mechanism. Materials and Methods Differential miRNA expression analysis was performed using next-generation sequencing (NGS) in the patients with PCa and benign prostatic hyperplasia (BPH). The expression levels of miR-100-5p were detected using real-time fluorescence quantitative PCR (qRT-PCR). PCa cells were transfected with NC-mimics or miR-100-5p mimics, inhibitor by using liposome transfection. Moreover, the CCK-8 proliferation assay, colony formation assay, cell scratch assay and Transwell assay were used to detect the effects of miR-100-5p on cell proliferation, migration, and invasion. In addition, the target gene of miR-100-5p was verified by luciferase reporter gene assay, and the influence of miR-100-5p on the expression of mTOR mRNA by qRT-PCR and the expression of mammalian target of rapamycin (mTOR) protein was detected by western blot and immunohistochemical staining. Results Differential expression analysis of high-throughput sequencing data showed low expression of miR-100-5p in the patients of PCa. It was further confirmed by qRT-PCR that the expression of miR-100-5p in PCa cells was significantly lower than that in RWPE-1 cells (P<0.01). miR-100-5p expression in lymph node carcinoma of prostate(LNCaP) cells was markedly upregulated after transfection with miR-100-5p mimics (P<0.01), while cell proliferation, migration and invasion capacities were clearly reduced (P<0.01). mTOR mRNA and protein expression was also substantially lowered (P<0.01) and mTOR adjusted the expression of NOX4. Finally, we further confirmed by immunohistochemical staining that miR-100-5p regulated the expression of mTOR and NOX4. Conclusion miR-100-5p is expressed at low levels in PCa cells, and it can suppress PCa cell proliferation, migration and invasion, the mechanism of which is related to downregulating the expression of mTOR.
Collapse
Affiliation(s)
- Yun Ye
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Su-Liang Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jian-Jun Wang
- Emergency Department, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
32
|
Zarà M, Amadio P, Campodonico J, Sandrini L, Barbieri SS. Exosomes in Cardiovascular Diseases. Diagnostics (Basel) 2020; 10:E943. [PMID: 33198302 PMCID: PMC7696149 DOI: 10.3390/diagnostics10110943] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Exosomes are nano-sized biovesicles of endocytic origin physiologically released by nearly all cell types into surrounding body fluids. They carry cell-specific cargos of protein, lipids, and genetic materials and can be selectively taken up by neighboring or distant cells. Since the intrinsic properties of exosomes are strictly influenced by the state of the parental cell and by the cellular microenvironment, the analysis of exosome origin and content, and their cell-targeting specificity, make them attractive as possible diagnostic and prognostic biomarkers. While the possible role of exosomes as messengers and a regenerative tool in cardiovascular diseases (CVDs) is actively investigated, the evidence about their usefulness as biomarkers is still limited and incomplete. Further complications are due to the lack of consensus regarding the most appropriate approach for exosome isolation and characterization, both important issues for their effective clinical translation. As a consequence, in this review, we will discuss the few information currently accessible about the diagnostic/prognostic potential of exosomes in CVDs and on the methodologies available for exosome isolation, analysis, and characterization.
Collapse
Affiliation(s)
- Marta Zarà
- Unit of Brain-Heart axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, via Parea 4, 20138 Milan, Italy; (P.A.); (L.S.)
| | - Patrizia Amadio
- Unit of Brain-Heart axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, via Parea 4, 20138 Milan, Italy; (P.A.); (L.S.)
| | - Jeness Campodonico
- Intensive Cardiac Care Unit, Centro Cardiologico Monzino IRCCS, via Parea 4, 20138 Milan, Italy;
| | - Leonardo Sandrini
- Unit of Brain-Heart axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, via Parea 4, 20138 Milan, Italy; (P.A.); (L.S.)
| | - Silvia S. Barbieri
- Unit of Brain-Heart axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, via Parea 4, 20138 Milan, Italy; (P.A.); (L.S.)
| |
Collapse
|
33
|
Zhang TR, Huang WQ. Angiogenic Exosome-Derived microRNAs: Emerging Roles in Cardiovascular Disease. J Cardiovasc Transl Res 2020; 14:824-840. [PMID: 33104961 DOI: 10.1007/s12265-020-10082-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Angiogenesis is the process of growing endothelial capillary cells. Exosomes are extracellular vesicles that are rich in miRNAs. Studies have shown that exosomes can carry communication between cells and various tissues by delivering miRNAs to their target organs and cells. It has been repeatedly proven that miRNAs regulate the expression of growth factors and other proteins in endothelial cells through paracrine signalling and participate in the physiological and pathological processes of angiogenesis. In the diagnosis and treatment of diseases, exosome-derived microRNAs can play important roles as biomarkers and drug carriers. In this review, we introduce the characteristics of miRNAs and exosomes and their interactions. Then, we specifically summarize the exosome-derived miRNAs related to angiogenesis, and we discuss the potential uses of exosome-derived miRNAs for diagnosing and treating cardiovascular diseases. Graphical abstract.
Collapse
Affiliation(s)
- Tian-Rong Zhang
- Department of Geriatric Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Disease Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, The Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Wei-Qiang Huang
- Department of Geriatric Cardiology & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Disease Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning, 530021, The Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
34
|
In vitro controlled release of extracellular vesicles for cardiac repair from poly(glycerol sebacate) acrylate-based polymers. Acta Biomater 2020; 115:92-103. [PMID: 32814141 DOI: 10.1016/j.actbio.2020.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
Cell therapy to restore cardiac function in chronic heart failure has been extensively studied. However, its therapeutic value is limited due to poor cell engraftment and survival and the therapeutic outcomes have been attributed to paracrine secretions such as extracellular vesicles (EV). The direct use of EV is an attractive therapeutic strategy and it has been shown that the kinetics of delivery of the EV to the targeted tissue may impact the outcomes. However, there are currently no technologies to deliver EV to the heart in a controlled and tunable manner. The objective of this study was to design a controlled release system, based on a photocurable adhesive polymer, to locally deliver EV to the cardiac tissue. We have first demonstrated that the adhesive polymer, PGSA-g-EG, did not impact the EV bioactivity in vitro and was biocompatible in vivo when tested in a rat model. Importantly, the polymer remained attached to the heart surface for at least 1 month. We have then evaluated and optimized the in vitro release kinetics of the EV from the PGSA-g-EG polymer. Freeze-dried EV formulations were developed to tune the release kinetics and maximize the loading in the polymeric material. Moreover, despite the instability of the EV in aqueous medium at 37°C, the PGSA-g-EG polymer was able to release bioactive EV for at least 14 days. Overall, these results suggest that the PGSA-g-EG is a suitable material to promote the controlled delivery of bioactive EV over an extended period of time. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EV) are an investigational class of therapeutics that has shown promise to restore cardiac function following an ischemic event. Furthermore, its translation to the clinics is expected to pose less regulatory challenges than cell-based therapies. However, EV therapeutic outcomes are likely to be impacted by the route of administration and the kinetics of delivery to the target tissue. Therefore, there is a need for biomaterial-based technologies to deliver, in a controlled and tunable manner, EV to the heart. The present study describes the use of PGSA-g-EG polymer as an adhesive cardiac patch with potential to enable the controlled delivery of bioactive EV over an extended period of time to the cardiac tissue.
Collapse
|
35
|
Chen Z, Yan Y, Wu J, Qi C, Liu J, Wang J. Expression level and diagnostic value of exosomal NEAT1/miR-204/MMP-9 in acute ST-segment elevation myocardial infarction. IUBMB Life 2020; 72:2499-2507. [PMID: 32916037 DOI: 10.1002/iub.2376] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/21/2022]
Abstract
Acute myocardium infarction (AMI) is one of the main causes of cardiovascular death, and timely intervention and diagnosis are essential. Owing to the irreversible apoptosis and death of myocardial cells, which ultimately causes heart failure, the problem of myocardial repair after myocardial infarction needs to be urgently addressed. Exosomes can act as messengers between cells, delivering large amounts of proteins, RNA, and lipids to receptor cells, and regulating target cell functions. Studies have shown that exosomes can repair infarcted myocardium. We aimed to investigate the relationship between long non-coding RNA NEAT1 in serum exosomes of patients and AMI and its underlying mechanism. Subjects were divided into control, UA, and STEMI groups. RNA was extracted from the serum exosomes, and the expressions of lncRNA NEAT1 and miR-204 were detected by qRT-PCR. MMP-9 was detected by western blot, Spearman test was used to analyze the correlation among the three. Logistic regression and Receiver-operating characteristic curve (ROC) were used to evaluate the prediction of acute myocardial infarction. The expressions of NEAT1 and MMP-9 in serum exosomes of patients with acute ST-segment elevation myocardial infarction were up-regulated and positively correlated, miR-204 expression was down-regulated, there were no correlations between miR-204 with NEAT1, or MMP-9. Exosomal NEAT1, miR-204, and MMP-9 displayed potent biomarkers for diagnosis of acute ST-segment elevation myocardial infarction.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junduo Wu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chao Qi
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
36
|
Established and Emerging Mechanisms in the Pathogenesis of Arrhythmogenic Cardiomyopathy: A Multifaceted Disease. Int J Mol Sci 2020; 21:ijms21176320. [PMID: 32878278 PMCID: PMC7503882 DOI: 10.3390/ijms21176320] [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: 08/11/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable myocardial disease that manifests with cardiac arrhythmias, syncope, sudden cardiac death, and heart failure in the advanced stages. The pathological hallmark of ACM is a gradual replacement of the myocardium by fibroadiposis, which typically starts from the epicardium. Molecular genetic studies have identified causal mutations predominantly in genes encoding for desmosomal proteins; however, non-desmosomal causal mutations have also been described, including genes coding for nuclear proteins, cytoskeleton componentsand proteins involved in excitation-contraction coupling. Despite the poor prognosis, currently available treatments can only partially control symptoms and to date there is no effective therapy for ACM. Inhibition of the canonical Wnt/β-catenin pathway and activation of the Hippo and the TGF-β pathways have been implicated in the pathogenesis of ACM. Yet, our understanding of the molecular mechanisms involved in the development of the disease and the cell source of fibroadiposis remains incomplete. Elucidation of the pathogenesis of the disease could facilitate targeted approaches for treatment. In this manuscript we will provide a comprehensive review of the proposed molecular and cellular mechanisms of the pathogenesis of ACM, including the emerging evidence on abnormal calcium homeostasis and inflammatory/autoimmune response. Moreover, we will propose novel hypothesis about the role of epicardial cells and paracrine factors in the development of the phenotype. Finally, we will discuss potential innovative therapeutic approaches based on the growing knowledge in the field.
Collapse
|
37
|
Zeng Q, Zhou Y, Liang D, He H, Liu X, Zhu R, Zhang M, Luo X, Wang Y, Huang G. Exosomes Secreted From Bone Marrow Mesenchymal Stem Cells Attenuate Oxygen-Glucose Deprivation/Reoxygenation-Induced Pyroptosis in PC12 Cells by Promoting AMPK-Dependent Autophagic Flux. Front Cell Neurosci 2020; 14:182. [PMID: 32765221 PMCID: PMC7379865 DOI: 10.3389/fncel.2020.00182] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Cerebral ischemia–reperfusion (I/R) injury can lead to severe dysfunction, and its treatment is difficult. It is reported that nucleotide-binding domain and leucine-rich repeat family protein 3 (NLRP3) inflammasome-mediated cell pyroptosis is an important part of cerebral I/R injury and the activation of autophagy can inhibit pyroptosis in some tissue injury. Our previous study found that the protective effects of bone marrow mesenchymal stem cells (BMSCs) in cerebral I/R injury may be associated with the regulation of autophagy. Recent studies have demonstrated that exosomes secreted from BMSCs (BMSC-Exos) may play an essential role in the effective biological performance of BMSCs and the protective mechanism of BMSC-Exos is associated with the activation of autophagy and the remission of inflammation, but it has not been reported in studies of cerebral I/R injury. We aimed to investigate the effects of BMSC-Exos on cerebral I/R injury and determine if the mechanism is associated with the regulation of pyroptosis and autophagic flux. Method: PC12 cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to induce cerebral I/R in vitro and were cocultured with BMSC-Exos. Cell viability was determined with CCK-8 and lactate dehydrogenase (LDH) detection kits. Scanning electron microscopy (SEM), Hoechst 33342/propidium iodide (PI) double staining, 2′,7′-dichlorodihydrofluorescein diacetate assay, immunofluorescence, Western blot, and Enzyme-linked immunosorbent assay (ELISA) were used to detect cell pyroptosis. Furthermore, transmission electron microscopy (TEM), GFP-RFP-LC3 adenovirus transfection, and Western blot were used to detect autophagic flux and its influence on pyroptosis. Finally, coimmunoprecipitation was used to detect the binding interaction between NLRP3 and LC3. Results: BMSC-Exos increased cell viability in OGD/R. The inhibitory effect of BMSC-Exos on pyroptosis was comparable to the NLRP3 inhibitor MCC950 and was reversed by NLRP3 overexpression. Furthermore, BMSC-Exos promoted autophagic flux through the AMP-activated kinase (AMPK)/mammalian target of the rapamycin pathway, whereas chloroquine, AMPK silencing, and compound C blocked the inhibitory effect on pyroptosis. Conclusions: BMSC-Exos can protect PC12 cells against OGD/R injury via attenuation of NLRP3 inflammasome-mediated pyroptosis by promoting AMPK-dependent autophagic flux.
Collapse
Affiliation(s)
- Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| | - Yuqing Zhou
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| | - Donghui Liang
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - He He
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| | - Xiaoli Liu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| | - Rui Zhu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| | - Meimei Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, China.,Shenzhen Sanming Project Group, Spaulding Rehabilitation Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Yao Wang
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan'ao People's Hospital, Shenzhen, China
| | - Guozhi Huang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Rehabilitation Medical School, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Mancuso T, Barone A, Salatino A, Molinaro C, Marino F, Scalise M, Torella M, De Angelis A, Urbanek K, Torella D, Cianflone E. Unravelling the Biology of Adult Cardiac Stem Cell-Derived Exosomes to Foster Endogenous Cardiac Regeneration and Repair. Int J Mol Sci 2020; 21:E3725. [PMID: 32466282 PMCID: PMC7279257 DOI: 10.3390/ijms21103725] [Citation(s) in RCA: 20] [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: 04/04/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac remuscularization has been the stated goal of the field of regenerative cardiology since its inception. Along with the refreshment of lost and dysfunctional cardiac muscle cells, the field of cell therapy has expanded in scope encompassing also the potential of the injected cells as cardioprotective and cardio-reparative agents for cardiovascular diseases. The latter has been the result of the findings that cell therapies so far tested in clinical trials exert their beneficial effects through paracrine mechanisms acting on the endogenous myocardial reparative/regenerative potential. The endogenous regenerative potential of the adult heart is still highly debated. While it has been widely accepted that adult cardiomyocytes (CMs) are renewed throughout life either in response to wear and tear and after injury, the rate and origin of this phenomenon are yet to be clarified. The adult heart harbors resident cardiac/stem progenitor cells (CSCs/CPCs), whose discovery and characterization were initially sufficient to explain CM renewal in response to physiological and pathological stresses, when also considering that adult CMs are terminally differentiated cells. The role of CSCs in CM formation in the adult heart has been however questioned by some recent genetic fate map studies, which have been proved to have serious limitations. Nevertheless, uncontested evidence shows that clonal CSCs are effective transplantable regenerative agents either for their direct myogenic differentiation and for their paracrine effects in the allogeneic setting. In particular, the paracrine potential of CSCs has been the focus of the recent investigation, whereby CSC-derived exosomes appear to harbor relevant regenerative and reparative signals underlying the beneficial effects of CSC transplantation. This review focuses on recent advances in our knowledge about the biological role of exosomes in heart tissue homeostasis and repair with the idea to use them as tools for new therapeutic biotechnologies for "cell-less" effective cardiac regeneration approaches.
Collapse
Affiliation(s)
- Teresa Mancuso
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Antonella Barone
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Alessandro Salatino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Claudia Molinaro
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Fabiola Marino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| |
Collapse
|
39
|
He N, Zhang Y, Zhang S, Wang D, Ye H. Exosomes: Cell-Free Therapy for Cardiovascular Diseases. J Cardiovasc Transl Res 2020; 13:713-721. [PMID: 32333198 DOI: 10.1007/s12265-020-09966-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/04/2020] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases (CVDs) are an important cause of death and disease worldwide. Because injured cardiac tissue cannot be repaired itself, it is urgent to develop other alternate therapies. Stem cells can be differentiated into cardiomyocytes, endothelial cells, and vascular smooth muscle cells for the treatment of CVDs. Therefore, cell therapy has recently been considered a viable treatment option that can significantly improve cardiac function. Nonetheless, implanted stem cells rarely survive in the recipient heart, suggesting that the benefits of stem cell therapy may involve other mechanisms. Exosomes derived from stem cells have a myocardial protection function after myocardial injury, and may be a promising and effective therapy for CVDs. Here, we discuss the application and mechanism of exosomes derived from stem cells in the diagnosis and treatment of CVDs and provide evidence for the application of exosomes in CVDs. Graphical Abstract.
Collapse
Affiliation(s)
- Nana He
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Yuelin Zhang
- Department of Medicine, University of Ningbo, Ningbo, China
| | - Shun Zhang
- Department of Experimental Medical Science, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo, China
| | - Dongjuan Wang
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China
| | - Honghua Ye
- Department of Cardiology, HwaMei Hospital (previously named Ningbo No. 2 Hospital), University of Chinese Academy of Sciences, 41 Xibei Street, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
40
|
Firoozi S, Pahlavan S, Ghanian MH, Rabbani S, Barekat M, Nazari A, Pakzad M, Shekari F, Hassani SN, Moslem F, Lahrood FN, Soleimani M, Baharvand H. Mesenchymal stem cell-derived extracellular vesicles alone or in conjunction with a SDKP-conjugated self-assembling peptide improve a rat model of myocardial infarction. Biochem Biophys Res Commun 2020; 524:903-909. [PMID: 32057366 DOI: 10.1016/j.bbrc.2020.02.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/01/2020] [Indexed: 01/02/2023]
Abstract
PURPOSE The aim of this study was to investigate the cardiac repair effect of human bone marrow mesenchymal stromal cells-derived extracellular vesicles (MSC-EVs) after intramyocardial injection in free form or encapsulated within a self-assembling peptide hydrogel modified with SDKP motif, in a rat model of myocardial infarction (MI). METHODS MSC-EVs were isolated by ultracentrifuge and characterized for physical parameters and surface proteins. Furthermore, cellular uptake and cardioprotective effects of MSC-EVs were evaluated in vitro using neonatal mouse cardiomyocytes (NMCMs). In vivo effects of MSC-EVs on cardiac repair were studied in rat MI model by comparing the vehicle group (injected with PBS), EV group (injected with MSC-EVs) and Gel + EV group (injected with MSC-EVs encapsulated in (RADA)4-SDKP hydrogel) with respect to cardiac function and fibrotic area using echocardiography and Masson's trichrome staining, respectively. Histological sections were assessed by α-SMA and CD68 immunostaining to investigate the angiogenic and anti-inflammatory effects of the MSC-EVs. RESULTS We observed the uptake of MSC-EVs into NMCMs which led to NMCMs protection against H2O2-induced oxidative stress by substantial reduction of apoptosis. In myocardial infarcted rats, cardiac function was improved after myocardial injection of MSC-EVs alone or in conjunction with (RADA)4-SDKP hydrogel. This functional restoration coincided with promotion of angiogenesis and decrement of fibrosis and inflammation. CONCLUSION These data demonstrated that MSC-EVs can be used alone as a potent therapeutic agent for improvement of myocardial infarction.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Animals, Newborn
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Biological Transport
- Biomarkers/metabolism
- Disease Models, Animal
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/transplantation
- Gene Expression
- Humans
- Hydrogels/administration & dosage
- Hydrogels/chemistry
- Hydrogen Peroxide/pharmacology
- Injections, Intramuscular
- Mesenchymal Stem Cells/chemistry
- Mesenchymal Stem Cells/cytology
- Mice
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/therapy
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Oxidative Stress
- Peptides/administration & dosage
- Primary Cell Culture
- Rats
Collapse
Affiliation(s)
- Saman Firoozi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad-Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahram Rabbani
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Barekat
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Abdoreza Nazari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Pakzad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fariba Moslem
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Nobakht Lahrood
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
41
|
Yang S, Zhu B, Yin P, Zhao L, Wang Y, Fu Z, Dang R, Xu J, Zhang J, Wen N. Integration of Human Umbilical Cord Mesenchymal Stem Cells-Derived Exosomes with Hydroxyapatite-Embedded Hyaluronic Acid-Alginate Hydrogel for Bone Regeneration. ACS Biomater Sci Eng 2020; 6:1590-1602. [PMID: 33455380 DOI: 10.1021/acsbiomaterials.9b01363] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The treatment of bone defects has plagued clinicians. Exosomes, the naturally secreted nanovesicles by cells, exhibit great potential in bone defect regeneration to realize cell-free therapy. In this work, we successfully revealed that human umbilical cord mesenchymal stem cells-derived exosomes could effectively promote the proliferation, migration, and osteogenic differentiation of a murine calvariae preosteoblast cell line in vitro. Considering the long period of bone regeneration, to effectively exert the reparative effect of exosomes, we synthesized an injectable hydroxyapatite (HAP)-embedded in situ cross-linked hyaluronic acid-alginate (HA-ALG) hydrogel system to durably retain exosomes at the defect sites. Then, we combined the exosomes with the HAP-embedded in situ cross-linked HA-ALG hydrogel system to repair bone defects in rats in vivo. The results showed that the combination of exosomes and composite hydrogel could significantly enhance bone regeneration. Our experiment provides a new strategy for exosome-based therapy, which shows great potential in future tissue and organ repair.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Biao Zhu
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Peng Yin
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lisheng Zhao
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yizhu Wang
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhiguang Fu
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Ruijie Dang
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Juan Xu
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Jianjun Zhang
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ning Wen
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
42
|
Zhang AB, Peng YF, Jia JJ, Nie Y, Zhang SY, Xie HY, Zhou L, Zheng SS. Exosome-derived galectin-9 may be a novel predictor of rejection and prognosis after liver transplantation. J Zhejiang Univ Sci B 2020; 20:605-612. [PMID: 31168974 DOI: 10.1631/jzus.b1900051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute cellular rejection (ACR) remains a major concern after liver transplantation. Predicting and monitoring acute rejection by non-invasive methods are very important for guiding the use of immunosuppressive drugs. Many studies have shown that exosomes and their contents are potential biomarkers for various liver diseases. Here, we identify and validate the role of exosomes and galectin-9 in ACR after liver transplantation. Exosomes were isolated from three sets of paired patients, with and without ACR, and the proteins within the exosomes were isolated and identified. Candidate proteins were then validated using a tissue microarray containing resected liver samples from 73 ACR and 63 non-rejection patients. Finally, protein expression and clinical manifestations were included in Kaplan-Meier survival and Cox regression analyses. Circulating exosomes were isolated from ACR and non-rejection patients and characterized using transmission electron microscopy and western blotting for CD63/CD81. Western blotting experiments revealed higher levels of galectin-9 protein in circulating exosomes from ACR recipients. Immunohistochemical analysis of the tissue microarray showed that the expression of galectin-9 in resected liver was significantly higher in the ACR group than in the non-rejection group (P<0.05). Higher levels of galectin-9 expression in resected livers were associated with poorer prognosis (P<0.05). Exosome-derived galectin-9 may be a novel predictor of rejection and prognosis after liver transplantation.
Collapse
Affiliation(s)
- Ai-Bin Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yi-Fan Peng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, the First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Jun-Jun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yu Nie
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Shi-Yu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, the First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Hai-Yang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, the First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, the First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| | - Shu-Sen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, the First Affiliated Hospital, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
43
|
Lin Y, Lu Y, Li X. Biological characteristics of exosomes and genetically engineered exosomes for the targeted delivery of therapeutic agents. J Drug Target 2019; 28:129-141. [PMID: 31280623 DOI: 10.1080/1061186x.2019.1641508] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A primary focus of pharmacology is the accurate transport of drugs from the peripheral veins and their delivery to specific tissues and organs. Exosomes are nanoscale extracellular vesicles with comparatively enhanced circulation stability, biocompatibility, physicochemical stability and bio-barrier permeation ability, as well as reduced toxicity. Therefore, they are considered a superior drug delivery platform. Core ligands and homing peptides fuse with transmembrane proteins on the exosome surface. Genetically engineered exosomes target specific tissues or organs and agents such as siRNA, miRNA and chemotherapeutics can be loaded into exosomes to improve the regulation of target tissues and organs. Here, we review exosome biogenesis, release, uptake and isolation. We also summarise the current applications of genetically engineered exosomes for tumours, and neurological, cardiovascular and liver diseases.
Collapse
Affiliation(s)
- Yan Lin
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China
| | - Yaqiong Lu
- Gansu Provincial Cancer Hospital, Gansu Provincial Academic Institute for Medical Research, Lanzhou, People's Republic of China
| | - Xun Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China.,The Fifth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, People's Republic of China.,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, People's Republic of China.,Hepatopancreatobiliary Surgery Institute of Gansu Province, Medical College Cancer Center of Lanzhou, Lanzhou, People's Republic of China
| |
Collapse
|
44
|
Harrell CR, Fellabaum C, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Molecular Mechanisms Responsible for Therapeutic Potential of Mesenchymal Stem Cell-Derived Secretome. Cells 2019; 8:cells8050467. [PMID: 31100966 PMCID: PMC6562906 DOI: 10.3390/cells8050467] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cell (MSC)-sourced secretome, defined as the set of MSC-derived bioactive factors (soluble proteins, nucleic acids, lipids and extracellular vesicles), showed therapeutic effects similar to those observed after transplantation of MSCs. MSC-derived secretome may bypass many side effects of MSC-based therapy, including unwanted differentiation of engrafted MSCs. In contrast to MSCs which had to be expanded in culture to reach optimal cell number for transplantation, MSC-sourced secretome is immediately available for treatment of acute conditions, including fulminant hepatitis, cerebral ischemia and myocardial infarction. Additionally, MSC-derived secretome could be massively produced from commercially available cell lines avoiding invasive cell collection procedure. In this review article we emphasized molecular and cellular mechanisms that were responsible for beneficial effects of MSC-derived secretomes in the treatment of degenerative and inflammatory diseases of hepatobiliary, respiratory, musculoskeletal, gastrointestinal, cardiovascular and nervous system. Results obtained in a large number of studies suggested that administration of MSC-derived secretomes represents a new, cell-free therapeutic approach for attenuation of inflammatory and degenerative diseases. Therapeutic effects of MSC-sourced secretomes relied on their capacity to deliver genetic material, growth and immunomodulatory factors to the target cells enabling activation of anti-apoptotic and pro-survival pathways that resulted in tissue repair and regeneration.
Collapse
Affiliation(s)
| | | | - Nemanja Jovicic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia.
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland.
| | - Nebojsa Arsenijevic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia.
| | - Vladislav Volarevic
- Department for Microbiology and Immunology, Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia.
| |
Collapse
|
45
|
Liu H, Sun X, Gong X, Wang G. Human umbilical cord mesenchymal stem cells derived exosomes exert antiapoptosis effect via activating PI3K/Akt/mTOR pathway on H9C2 cells. J Cell Biochem 2019; 120:14455-14464. [PMID: 30989714 DOI: 10.1002/jcb.28705] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/03/2019] [Accepted: 03/15/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Hui Liu
- Department of Cardiology, State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases Chinese Academy of Medical Sciences and Peking Union Medical College Beijing People's Republic of China
| | - Xiaolu Sun
- Department of Cardiology, State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases Chinese Academy of Medical Sciences and Peking Union Medical College Beijing People's Republic of China
| | - Xuhe Gong
- Department of Cardiology Beijing Friendship Hospital, Capital Medical University Beijing People's Republic of China
| | - Guogan Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases Chinese Academy of Medical Sciences and Peking Union Medical College Beijing People's Republic of China
| |
Collapse
|
46
|
Wu R, Gao W, Yao K, Ge J. Roles of Exosomes Derived From Immune Cells in Cardiovascular Diseases. Front Immunol 2019; 10:648. [PMID: 30984201 PMCID: PMC6449434 DOI: 10.3389/fimmu.2019.00648] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/11/2019] [Indexed: 12/21/2022] Open
Abstract
Therapies aimed at minimizing adverse remodeling in cardiovascular diseases on a molecular and cellular basis are urgently needed. Exosomes are nanosized lipid vesicles released from various cells that are able to mediate intercellular signaling and communication via their cargos. It has been increasingly demonstrated that exosomes from cardiomyocytes or stem/progenitor cells can promote cardiac repair and regeneration, but their mechanism has not been fully explained. Immune responses mediated by immune cells also play important and complicated roles in the progression of various cardiovascular diseases such as myocardial infarction and atherosclerosis. Exosomes derived from immune cells have shown pleiotropic effects on these pathological states, whether similar to or different from their parent cells. However, the underlying mechanism remains obscure. In this review, we first describe the biological characteristics and biogenesis of exosomes. Then we critically examine the emerging roles of exosomes in cardiovascular disease; the exosomes we focus on are derived from immune cells such as dendritic cells, macrophages, B cells, T cells, as well as neutrophils and mast cells. Among the cardiovascular diseases we discuss, we mainly focus on myocardial infarction and atherosclerosis. As active intercellular communicators, exosomes from immune cells may offer prospective diagnostic and therapeutic value in cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Kang Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| |
Collapse
|
47
|
Bone marrow mesenchymal stem cell-derived exosomes alleviate high phosphorus-induced vascular smooth muscle cells calcification by modifying microRNA profiles. Funct Integr Genomics 2019; 19:633-643. [PMID: 30850904 DOI: 10.1007/s10142-019-00669-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/04/2019] [Accepted: 02/15/2019] [Indexed: 12/19/2022]
Abstract
Vascular calcification is a common complication in patients with chronic kidney disease (CKD). It is an important predictor of cardiovascular disease and all-cause mortality. Previous studies have confirmed that bone marrow mesenchymal stem cell (BMSC) therapy can reduce vascular calcification, but the specific mechanism is still controversial. In this study, we aimed to investigate the mechanisms of BMSC-derived exosomes (EXO) in improving vascular calcification. BMSCs were cultured and EXO were isolated using the Total Exosome Isolation Reagent. Human aortic vascular smooth muscle cells (HA-VSMCs) were cultured into three groups: control group, high phosphorus group, and high phosphorus plus EXO group. Then, indicators related to smooth muscle cell calcification and microRNA profiles were analyzed. BMSC-derived exosomes inhibited high phosphorus-induced calcification in HA-VSMCs. Besides, EXO treatment reduced calcium content and decreased the alkaline phosphatase (AKP) activity in high phosphorus co-incubated HA-VSMCs. MicroRNA (miRNA) and mRNA expression profiles analyses revealed that 63 miRNAs were significantly upregulated and 1424 genes were significantly downregulated in HA-VSMCs after EXO treatment. Functional miRNA-gene regulatory network revealed that mTOR, MAPK, and Wnt signaling pathway were involved in vascular calcification. BMSC-derived exosomes alleviated high phosphorus-induced calcification in HA-VSMC through modifying miRNA profiles.
Collapse
|
48
|
Li SL, An N, Liu B, Wang SY, Wang JJ, Ye Y. Exosomes from LNCaP cells promote osteoblast activity through miR-375 transfer. Oncol Lett 2019; 17:4463-4473. [PMID: 30988815 PMCID: PMC6447935 DOI: 10.3892/ol.2019.10110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023] Open
Abstract
Previous studies have revealed that exosomes influence tumour metastasis, diagnosis and treatment. In addition, exosomal microRNAs (miRNAs/miRs) are closely associated with the metastatic microenvironment; however, the regulatory role of exosomal miRNAs from prostate cancer cells on bone metastasis remains poorly understood. In the present study, a series of experiments were performed to determine whether exosomal miR-375 from LNCaP cells promote osteoblast activity. Exosomes were isolated and purified by ultracentrifugation, total RNA from cells and total miRNA from exosomes were then extracted, and miR-375 levels were detected by reverse transcription-quantitative polymerase chain reaction. Exosome libraries from LNCaP and RWPE-1 cells were sequenced and selected using an Illumina HiSeq™ 2500 system. The effects of exosomes on osteoblasts were determined and osteoblast activity was evaluated by measuring the activity of alkaline phosphatase, the extent of extracellular matrix mineralisation and the expression of osteoblast activity-associated marker genes. Morphological observations, particle size analysis and molecular phenotyping confirmed that cell supernatants contained exosomes. Differential expression analysis confirmed high miR-375 expression levels in LNCaP cell-derived exosomes. The ability of exosomes to enter osteoblasts and increase their levels of miR-375 was further analysed. The results demonstrated that exosomal miR-375 significantly promoted osteoblast activity. In conclusion, the present study may lead to further investigation of the function role of exosomal miR-375 in the activation and differentiation of osteoblasts in PCa.
Collapse
Affiliation(s)
- Su-Liang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Na An
- Department of Laboratory Medicine, Shaanxi Jiaotong Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Bing Liu
- Department of Pathology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Sheng-Yu Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Jian-Jun Wang
- Intensive Care Unit, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| | - Yun Ye
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, P.R. China
| |
Collapse
|
49
|
Cho HM, Cho JY. Prediction of therapeutic effects of human cardiomyocytes in myocardial infarction using non-human primates model. ANNALS OF TRANSLATIONAL MEDICINE 2019; 6:S64. [PMID: 30613639 DOI: 10.21037/atm.2018.10.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hyun-Min Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
50
|
|