1
|
Watanabe T, Hatayama N, Guo M, Yuhara S, Shinoka T. Bridging the Gap: Advances and Challenges in Heart Regeneration from In Vitro to In Vivo Applications. Bioengineering (Basel) 2024; 11:954. [PMID: 39451329 PMCID: PMC11505552 DOI: 10.3390/bioengineering11100954] [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: 08/06/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 10/26/2024] Open
Abstract
Cardiovascular diseases, particularly ischemic heart disease, area leading cause of morbidity and mortality worldwide. Myocardial infarction (MI) results in extensive cardiomyocyte loss, inflammation, extracellular matrix (ECM) degradation, fibrosis, and ultimately, adverse ventricular remodeling associated with impaired heart function. While heart transplantation is the only definitive treatment for end-stage heart failure, donor organ scarcity necessitates the development of alternative therapies. In such cases, methods to promote endogenous tissue regeneration by stimulating growth factor secretion and vascular formation alone are insufficient. Techniques for the creation and transplantation of viable tissues are therefore highly sought after. Approaches to cardiac regeneration range from stem cell injections to epicardial patches and interposition grafts. While numerous preclinical trials have demonstrated the positive effects of tissue transplantation on vasculogenesis and functional recovery, long-term graft survival in large animal models is rare. Adequate vascularization is essential for the survival of transplanted tissues, yet pre-formed microvasculature often fails to achieve sufficient engraftment. Recent studies report success in enhancing cell survival rates in vitro via tissue perfusion. However, the transition of these techniques to in vivo models remains challenging, especially in large animals. This review aims to highlight the evolution of cardiac patch and stem cell therapies for the treatment of cardiovascular disease, identify discrepancies between in vitro and in vivo studies, and discuss critical factors for establishing effective myocardial tissue regeneration in vivo.
Collapse
Affiliation(s)
- Tatsuya Watanabe
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
| | - Naoyuki Hatayama
- Department of Anatomy, Aichi Medical University, Nagakute 480-1195, Japan;
| | - Marissa Guo
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
- Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Satoshi Yuhara
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA; (T.W.); (M.G.); (S.Y.)
| | - Toshiharu Shinoka
- Department of Surgery, Ohio State University, Columbus, OH 43210, USA
- Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
2
|
Lee H, Cho HJ, Han Y, Lee SH. Mid- to long-term efficacy and safety of stem cell therapy for acute myocardial infarction: a systematic review and meta-analysis. Stem Cell Res Ther 2024; 15:290. [PMID: 39256845 PMCID: PMC11389242 DOI: 10.1186/s13287-024-03891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/21/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND This comprehensive systematic review and meta-analysis investigated the mid- to long-term efficacy and safety of stem cell therapy in patients with acute myocardial infarction (AMI). METHODS The study encompassed 79 randomized controlled trials with 7103 patients, rendering it the most up-to-date and extensive analysis in this field. This study specifically focused on the impact of stem cell therapy on left ventricular ejection fraction (LVEF), major adverse cardiac events (MACE), and infarct size. RESULTS Stem cell therapy significantly improved LVEF at 6, 12, 24, and 36 months post-transplantation compared to control values, indicating its potential for long-term cardiac function enhancement. A trend toward reduced MACE occurrence was observed in the intervention groups, suggesting the potential of stem cell therapy to lower the risk of cardiovascular death, reinfarction, and stroke. Significant LVEF improvements were associated with long cell culture durations exceeding 1 week, particularly when combined with high injected cell quantities (at least 108 cells). No significant reduction in infarct size was observed. CONCLUSIONS This review highlights the potential of stem cell therapy as a promising therapeutic approach for patients with AMI, offering sustained LVEF improvement and a potential reduction in MACE risk. However, further research is required to optimize cell culture techniques, determine the optimal timing and dosage, and investigate procedural variations to maximize the efficacy and safety of stem cell therapy in this context.
Collapse
Affiliation(s)
- Hyeongsuk Lee
- College of Nursing, Research Institute of AI and Nursing Science, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, South Korea
| | - Hyun-Jai Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yeonjung Han
- College of Nursing, Research Institute of AI and Nursing Science, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, South Korea
| | - Seon Heui Lee
- College of Nursing, Research Institute of AI and Nursing Science, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon, 21936, South Korea.
| |
Collapse
|
3
|
Hosseinpour A, Hosseinpour H, Attar A. Preventive Effect of Bone Marrow Mononuclear Cell Transplantation on Acute Myocardial Infarction-Induced Heart Failure: A Meta-analysis of Randomized Controlled Trials. Cardiovasc Drugs Ther 2023; 37:1143-1153. [PMID: 35876933 DOI: 10.1007/s10557-022-07359-3] [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] [Accepted: 06/11/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Heart failure (HF) is a major complication of acute myocardial infarction (AMI). Transplantation of bone marrow mononuclear cells (BM-MNC) in the setting of AMI has been proposed as a means for myocardial tissue regeneration. Several trials have explored the outcomes of these cells on surrogate end points such as left ventricular ejection fraction (LVEF) in patients with AMI. However, the data regarding the clinical efficacy are infrequent. Here, we performed a meta-analysis investigating the effect of BM-MNCs injection on the rate of hospitalization for HF in the long-term follow-up period. METHODS PubMed, Scopus, and Cochrane databases were queried with various combinations of keywords through May 2, 2022. A random-effects meta-analysis was performed to calculate risk ratio (RR) and 95% confidence interval (CI) of hospitalization for HF, all-cause mortality, and stroke rate. Subgroup analyses for hospitalization based on time and cell dose were performed. RESULTS A total of 2150 patients with AMI across 22 trials were included for quantitative synthesis. At long-term follow-up, AMI patients treated with an intracoronary injection of BM-MNCs were less likely to be hospitalized for heart failure compared to the control group receiving standard treatment (RR = 0.54, 95% CI = [0.37; 0.78], p = 0.002). There was no association between BM-MNC therapy and all-cause mortality (RR = 0.69, 95% CI = [0.47; 1.01], p = 0.05) and stroke (RR = 1.12, 95% CI= [0.24; 5.21], p = 0.85). CONCLUSION Autologous injection of BM-MNC in the setting of AMI may be associated with decreased risk of hospitalization of heart failure in the long term. However, its effect on all-cause mortality and stroke rate is questionable.
Collapse
Affiliation(s)
- Alireza Hosseinpour
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Zand Street, Shiraz, Iran
| | | | - Armin Attar
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Zand Street, Shiraz, Iran.
| |
Collapse
|
4
|
Pezhouman A, Nguyen NB, Kay M, Kanjilal B, Noshadi I, Ardehali R. Cardiac regeneration - Past advancements, current challenges, and future directions. J Mol Cell Cardiol 2023; 182:75-85. [PMID: 37482238 DOI: 10.1016/j.yjmcc.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity worldwide. Despite improvements in the standard of care for patients with heart diseases, including innovation in pharmacotherapy and surgical interventions, none have yet been proven effective to prevent the progression to heart failure. Cardiac transplantation is the last resort for patients with severe heart failure, but donor shortages remain a roadblock. Cardiac regenerative strategies include cell-based therapeutics, gene therapy, direct reprogramming of non-cardiac cells, acellular biologics, and tissue engineering methods to restore damaged hearts. Significant advancements have been made over the past several decades within each of these fields. This review focuses on the advancements of: 1) cell-based cardiac regenerative therapies, 2) the use of noncoding RNA to induce endogenous cell proliferation, and 3) application of bioengineering methods to promote retention and integration of engrafted cells. Different cell sources have been investigated, including adult stem cells derived from bone marrow and adipose cells, cardiosphere-derived cells, skeletal myoblasts, and pluripotent stem cells. In addition to cell-based transplantation approaches, there have been accumulating interest over the past decade in inducing endogenous CM proliferation for heart regeneration, particularly with the use of noncoding RNAs such as miRNAs and lncRNAs. Bioengineering applications have focused on combining cell-transplantation approaches with fabrication of a porous, vascularized scaffold using biomaterials and advanced bio-fabrication techniques that may offer enhanced retention of transplanted cells, with the hope that these cells would better engraft with host tissue to improve cardiac function. This review summarizes the present status and future challenges of cardiac regenerative therapies.
Collapse
Affiliation(s)
- Arash Pezhouman
- Baylor College of Medicine, Department of Medicine, Division of Cardiology, Houston, Texas 77030, United States; Texas Heart Institute, Houston, Texas 77030, United States
| | - Ngoc B Nguyen
- Baylor College of Medicine, Department of Internal Medicine, Houston, Texas 77030, United States
| | - Maryam Kay
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA 90095, United States
| | - Baishali Kanjilal
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Iman Noshadi
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Reza Ardehali
- Baylor College of Medicine, Department of Medicine, Division of Cardiology, Houston, Texas 77030, United States; Texas Heart Institute, Houston, Texas 77030, United States.
| |
Collapse
|
5
|
Clavellina D, Balkan W, Hare JM. Stem cell therapy for acute myocardial infarction: Mesenchymal Stem Cells and induced Pluripotent Stem Cells. Expert Opin Biol Ther 2023; 23:951-967. [PMID: 37542462 PMCID: PMC10837765 DOI: 10.1080/14712598.2023.2245329] [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: 06/13/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) remains a leading cause of death in the United States. The limited capacity of cardiomyocytes to regenerate and the restricted contractility of scar tissue after AMI are not addressed by current pharmacologic interventions. Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach due to their low antigenicity, ease of harvesting, and efficacy and safety in preclinical and clinical studies, despite their low survival and engraftment rates. Other stem cell types, such as induced pluripotent stem cells (iPSCs) also show promise, and optimizing cardiac repair requires integrating emerging technologies and strategies. AREAS COVERED This review offers insights into advancing cell-based therapies for AMI, emphasizing meticulously planned trials with a standardized definition of AMI, for a bench-to-bedside approach. We critically evaluate fundamental studies and clinical trials to provide a comprehensive overview of the advances, limitations and prospects for cell-based therapy in AMI. EXPERT OPINION MSCs continue to show potential promise for treating AMI and its sequelae, but addressing their low survival and engraftment rates is crucial for clinical success. Integrating emerging technologies such as pluripotent stem cells and conducting well-designed trials will harness the full potential of cell-based therapy in AMI management. Collaborative efforts are vital to developing effective stem cell therapies for AMI patients.
Collapse
Affiliation(s)
- Diana Clavellina
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
6
|
Davidson SJ, Roncalli J, Surder D, Corti R, Chugh AR, Yang PC, Henry TD, Stanberry L, Lemarchand P, Beregi JP, Traverse JH. Microvascular obstruction identifies a subgroup of patients who benefit from stem cell therapy following ST-elevation myocardial infarction. Am Heart J 2023; 259:79-86. [PMID: 36796572 DOI: 10.1016/j.ahj.2023.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/01/2023] [Accepted: 02/05/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Microvascular obstruction (MVO) is associated with greater infarct size, adverse left-ventricular (LV) remodeling and reduced ejection fraction following ST-elevation myocardial infarction (STEMI). We hypothesized that patients with MVO may constitute a subgroup of patients that would benefit from intracoronary stem cell delivery with bone marrow mononuclear cells (BMCs) given previous findings that BMCs tended to improve LV function only in patients with significant LV dysfunction. METHODS AND RESULTS We analyzed the cardiac MRIs of 356 patients (303 M, 53 F) with anterior STEMIs who received autologous BMCs or placebo / control as part of 4 randomized clinical trials that included the Cardiovascular Cell Therapy Research Network (CCTRN) TIME trial and its pilot, the multicenter French BONAMI trial and SWISS-AMI trials. A total of 327 patients had paired imaging data at 1 year. All patients received 100 to 150 million intracoronary autologous BMCs or placebo / control 3 to 7 days following primary PCI and stenting. LV function, volumes, infarct size and MVO were assessed prior to infusion of BMCs and 1 year later. Patients with MVO (n = 210) had reduced LVEF and much greater infarct size and LV volumes compared to patients without MVO (n = 146) (P < .01). At 12 months, patients with MVO who received BMCs had significantly greater recovery of LVEF compared to those patients with MVO who received placebo (absolute difference = 2.7%; P < .05). Similarly, left-ventricular end-diastolic (LVEDVI) and end-systolic volume indices (LVESVI) demonstrated significantly less adverse remodeling in patients with MVO who received BMCs compared to placebo. In contrast, no improvement in LVEF or LV volumes was observed in those patients without MVO who received BMCs compared to placebo. CONCLUSIONS The presence of MVO on cardiac MRI following STEMI identifies a subgroup of patients who benefit from intracoronary stem cell therapy.
Collapse
Affiliation(s)
| | - Jerome Roncalli
- Federation de Cardiologie, Departmentie, Institute CARDIOMET, University Hospital of Toulouse, Toulose, France
| | - Daniel Surder
- Department of Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich Switzerland
| | - Roberto Corti
- Department of Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich Switzerland
| | - Atul R Chugh
- Franciscan Health Indiana Heart Physicians, Indianapolis, IN
| | | | | | - Larissa Stanberry
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN
| | - Patricia Lemarchand
- Institut du thorax, INSERM, CNRS, Universite de Nantes, CHU Nantes, Nantes, France
| | - Jeau-Paul Beregi
- Nimes Medical Imaging Group, University Montpellier, Nimes, France
| | - Jay H Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, Minneapolis, MN; University of Minnesota School of Medicine; Cardiovascular Division, Minneapolis, MN.
| |
Collapse
|
7
|
Kishino Y, Fukuda K. Unlocking the Pragmatic Potential of Regenerative Therapies in Heart Failure with Next-Generation Treatments. Biomedicines 2023; 11:biomedicines11030915. [PMID: 36979894 PMCID: PMC10046277 DOI: 10.3390/biomedicines11030915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Patients with chronic heart failure (HF) have a poor prognosis due to irreversible impairment of left ventricular function, with 5-year survival rates <60%. Despite advances in conventional medicines for HF, prognosis remains poor, and there is a need to improve treatment further. Cell-based therapies to restore the myocardium offer a pragmatic approach that provides hope for the treatment of HF. Although first-generation cell-based therapies using multipotent cells (bone marrow-derived mononuclear cells, mesenchymal stem cells, adipose-derived regenerative cells, and c-kit-positive cardiac cells) demonstrated safety in preclinical models of HF, poor engraftment rates, and a limited ability to form mature cardiomyocytes (CMs) and to couple electrically with existing CMs, meant that improvements in cardiac function in double-blind clinical trials were limited and largely attributable to paracrine effects. The next generation of stem cell therapies uses CMs derived from human embryonic stem cells or, increasingly, from human-induced pluripotent stem cells (hiPSCs). These cell therapies have shown the ability to engraft more successfully and improve electromechanical function of the heart in preclinical studies, including in non-human primates. Advances in cell culture and delivery techniques promise to further improve the engraftment and integration of hiPSC-derived CMs (hiPSC-CMs), while the use of metabolic selection to eliminate undifferentiated cells will help minimize the risk of teratomas. Clinical trials of allogeneic hiPSC-CMs in HF are now ongoing, providing hope for vast numbers of patients with few other options available.
Collapse
Affiliation(s)
| | - Keiichi Fukuda
- Correspondence: ; Tel.: +81-3-5363-3874; Fax: +81-3-5363-3875
| |
Collapse
|
8
|
Mahmud S, Alam S, Emon NU, Boby UH, Kamruzzaman, Ahmed F, Monjur-Al-Hossain ASM, Tahamina A, Rudra S, Ajrin M. Opportunities and challenges in stem cell therapy in cardiovascular diseases: Position standing in 2022. Saudi Pharm J 2022; 30:1360-1371. [PMID: 36249945 PMCID: PMC9563042 DOI: 10.1016/j.jsps.2022.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/17/2022] [Indexed: 10/29/2022] Open
Abstract
This study intends to evaluate the development, importance, pre-clinical and clinical study evaluation of stem cell therapy for the treatment of cardiovascular disease. Cardiovascular disease is one of the main causes of fatality in the whole world. Though there are great progressions in the pharmacological and other interventional treatment options, heart diseases remain a common disorder that causes long-term warnings. Recent accession promotes the symptoms and slows down the adverse effects regarding cardiac remodelling. But they cannot locate the problems of immutable loss of cardiac tissues. In this case, stem cell treatment holds a promising challenge. Stem cells are the cells that are capable of differentiating into many cells according to their needs. So, it is assumed that these cells can distinguish into many cells and if these cells can be individualized into cardiac cells then they can be used to replace the damaged tissues of the heart. There is some abridgment in this therapy, none the less stem cell therapy remains a hopeful destination in the treatment of heart disease.
Collapse
Affiliation(s)
- Shabnur Mahmud
- School of Health and Life Sciences, Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh
| | - Safaet Alam
- Pharmaceutical Sciences Research Division, BCSIR Laboratories, Dhaka, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dr. Qudrat-I-Khuda Road, Dhanmondi, Dhaka 1205, Bangladesh
| | - Nazim Uddin Emon
- Department of Pharmacy, Faculty of Science and Engineering, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Umme Habiba Boby
- Department of Pharmacy, Faculty of Science and Engineering, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Kamruzzaman
- Department of Pharmacy, Faculty of Science and Engineering, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Firoj Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka 1205, Bangladesh
| | - A S M Monjur-Al-Hossain
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka 1205, Bangladesh
| | - Afroza Tahamina
- Department of Botany, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Sajib Rudra
- Department of Botany, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Marzina Ajrin
- Department of Pharmacy, University of Science and Technology Chittagong, Chittagong 4202, Bangladesh
| |
Collapse
|
9
|
Mehta C, Shah R, Yanamala N, Sengupta PP. Cardiovascular Imaging Databases: Building Machine Learning Algorithms for Regenerative Medicine. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00216-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
10
|
Attar A, Hosseinpour A, Hosseinpour H, Kazemi A. Major cardiovascular events after bone marrow mononuclear cell transplantation following acute myocardial infarction: an updated post-BAMI meta-analysis of randomized controlled trials. BMC Cardiovasc Disord 2022; 22:259. [PMID: 35681123 PMCID: PMC9185901 DOI: 10.1186/s12872-022-02701-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/03/2022] [Indexed: 11/23/2022] Open
Abstract
Background The effect of bone marrow-derived mononuclear cells (BM-MNCs) after acute myocardial infarction (AMI) on myocardial function indices such as left ventricular ejection fraction has been widely studied. However, the effect of this intervention on major adverse cardiovascular events (MACE) was not the principal purpose of most investigations and its role is unclear. The aim of this study was to investigate the possible long-term clinical efficacy of BM-MNCs on MACE after AMI. Methods A comprehensive search was conducted through electronic databases for potentially eligible randomized trials investigating the impact of BM-MNC therapy following acute MI on clinical outcomes. Risk of bias of the eligible studies was assessed using the Cochrane Collaboration’s tool. The effect of treatment was displayed by risk ratio (RR) and its 95% confidence interval (CI) using random-effects model. Results Initial database searching found 1540 records and 23 clinical trials with a total of 2286 participants eligible for meta-analysis. Injection of BM-MNCs was associated with lower risk of composite end points of hospitalization for congestive heart failure (CHF), re-infarction, and cardiac-related mortality (91/1191 vs. 111/812, RR = 0.643, 95% CI = 0.489 to 0.845, p = 0.002). This effect was derived from both reduction of CHF (47/1220 vs. 62/841, RR = 0.568, 95% CI = 0.382 to 0.844, p = 0.005) and re-infarction rate (23/1159 vs. 30/775, RR = 0.583, 95% CI = 0.343 to 0.991, p = 0.046), but not cardiac-related mortality (28/1290 vs. 31/871, RR = 0.722, 95% CI = 0.436 to 1.197, p = 0.207). Conclusion This is the first meta-analysis focused on the cardiovascular outcomes of stem cell therapy after AMI and it revealed that transplantation of BM-MNCs may reduce composite endpoint of hospitalization for CHF, re-infarction, and cardiac related mortality driven mainly by reducing reinfarction and hospitalization for heart failure rates but not cardiovascular mortality. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02701-x.
Collapse
Affiliation(s)
- Armin Attar
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Alireza Hosseinpour
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Asma Kazemi
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Hosseinpour A, Kheshti F, Kazemi A, Attar A. Comparing the effect of bone marrow mono-nuclear cells with mesenchymal stem cells after acute myocardial infarction on improvement of left ventricular function: a meta-analysis of clinical trials. Stem Cell Res Ther 2022; 13:203. [PMID: 35578329 PMCID: PMC9109324 DOI: 10.1186/s13287-022-02883-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The effect of transplantation of bone-marrow mononuclear cells (BM-MNCs) and mesenchymal stem cells (MSCs) on ejection fraction (LVEF) has been studied in patients with acute myocardial infarction (AMI) in clinical trials. This raises the question that which type of cell may help improve LVEF better in AMI patients. No meta-analysis of clinical trials has yet addressed this question. METHODS Electronic databases were searched thoroughly to find eligible trials on the effects of transplantation of BM-MNCs and MSCs in patients with AMI. The primary outcome was improvement in LVEF. Data were synthesized using random-effects meta-analysis. For maximizing the credibility of subgroup analysis, we used the instrument for assessing the Credibility of Effect Modification of Analyses (ICEMAN) for meta-analyses. RESULTS A total of 36 trials (26 on BM-MNCs and 10 on MSCs) with 2489 patients (1466 were transplanted [1241 with BM-MNCs and 225 with MSCs] and 1023 as controls) were included. Both types of cells showed significant improvements in ejection fraction in short-term follow-up (BM-MNCs: WMD = 2.13%, 95% CI = 1.23 to 3.04, p < 0.001; MSCs: WMD = 3.71%, 95% CI = 2.32 to 5.09, p < 0.001), and according to ICEMAN criteria, MSCs are more effective. For selected population of patients who received stem cell transplantation in early course after AMI (less than 11 days), this effect was even more pronounced (BM-MNC: WMD = 3.07%, 95% CI = 1.97 to 4.17, p < 0.001, I2 = 40.7%; MSCs: WMD = 5.65%, 95% CI = 3.47 to 7.84, p < 0.001, I2 = 84.6%). CONCLUSION Our results showed that transplantation of MSCs after AMI might increase LVEF more than BM-MNCs; also, based on ICEMAN, there was likely effect modification between subgroups although uncertainty still remained.
Collapse
Affiliation(s)
- Alireza Hosseinpour
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Kheshti
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Kazemi
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armin Attar
- Department of Cardiovascular Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Apelin-13 Pretreatment Promotes the Cardioprotective Effect of Mesenchymal Stem Cells against Myocardial Infarction by Improving Their Survival. Stem Cells Int 2022; 2022:3742678. [PMID: 35355588 PMCID: PMC8960019 DOI: 10.1155/2022/3742678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/23/2022] Open
Abstract
Although mesenchymal stem cell- (MSC-) based therapy has shown promising results for myocardial infarction (MI), low cell survival heavily limits its beneficial effects. Apelin plays an essential regulatory role in cell proliferation. This study was aimed at determining whether Apelin-13 pretreatment could improve the survival of MSCs in the ischemic heart and enhance their cardioprotective efficacy against MI. MSCs were pretreated with or without Apelin-13 for 24 hours and then exposed to serum deprivation and hypoxia (SD/H) for 48 hours. The mitochondrial morphology of MSCs was assessed by MitoTracker staining. The apoptosis of MSCs was determined by TUNEL staining. The level of mitochondrial reactive oxygen species (ROS) of MSCs was detected by Mito-Sox staining. MSCs and Apelin-13-pretreated MSCs were transplanted into the peri-infarct region in a mouse MI model. Apelin-13 pretreatment protected MSCs against SD/H-induced mitochondrial fragmentation and apoptosis. Apelin-13 pretreatment reduced ROS generation induced by SD/H in MSCs. Furthermore, Apelin-13 pretreatment enhanced the angiogenesis of MSCs under SD/H conditions. Mechanistically, Apelin-13 pretreatment inhibited SD/H-induced MSC apoptosis by downregulating mitochondrial fission via activation of the ERK pathway, and these effects were partially abrogated by ERK inhibitor U0126. Apelin-13 pretreatment promoted the survival of MSCs in the ischemic heart. Moreover, transplantation with Apelin-13-pretreated MSCs improved heart function and increased angiogenesis accompanied by decreased fibrosis compared with MSC transplantation at 28 days following MI. These findings reveal that pretreatment with Apelin-13 improves MSCs survival and enhances their therapeutic efficacy for MI. Our study provides a novel approach to improve MSC-based therapy for cardiovascular disease.
Collapse
|
13
|
Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Ther 2022; 7:78. [PMID: 35273164 PMCID: PMC8913803 DOI: 10.1038/s41392-022-00925-z] [Citation(s) in RCA: 258] [Impact Index Per Article: 129.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Although the treatment of myocardial infarction (MI) has improved considerably, it is still a worldwide disease with high morbidity and high mortality. Whilst there is still a long way to go for discovering ideal treatments, therapeutic strategies committed to cardioprotection and cardiac repair following cardiac ischemia are emerging. Evidence of pathological characteristics in MI illustrates cell signaling pathways that participate in the survival, proliferation, apoptosis, autophagy of cardiomyocytes, endothelial cells, fibroblasts, monocytes, and stem cells. These signaling pathways include the key players in inflammation response, e.g., NLRP3/caspase-1 and TLR4/MyD88/NF-κB; the crucial mediators in oxidative stress and apoptosis, for instance, Notch, Hippo/YAP, RhoA/ROCK, Nrf2/HO-1, and Sonic hedgehog; the controller of myocardial fibrosis such as TGF-β/SMADs and Wnt/β-catenin; and the main regulator of angiogenesis, PI3K/Akt, MAPK, JAK/STAT, Sonic hedgehog, etc. Since signaling pathways play an important role in administering the process of MI, aiming at targeting these aberrant signaling pathways and improving the pathological manifestations in MI is indispensable and promising. Hence, drug therapy, gene therapy, protein therapy, cell therapy, and exosome therapy have been emerging and are known as novel therapies. In this review, we summarize the therapeutic strategies for MI by regulating these associated pathways, which contribute to inhibiting cardiomyocytes death, attenuating inflammation, enhancing angiogenesis, etc. so as to repair and re-functionalize damaged hearts.
Collapse
|
14
|
Botleroo RA, Bhandari R, Ahmed R, Kareem R, Gyawali M, Venkatesan N, Ogeyingbo OD, Elshaikh AO. Stem Cell Therapy for the Treatment of Myocardial Infarction: How Far Are We Now? Cureus 2021; 13:e17022. [PMID: 34522503 PMCID: PMC8425504 DOI: 10.7759/cureus.17022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
Myocardial infarction is one of the leading causes of death worldwide. Poor functional recovery of the myocardium is noticed after an event of myocardial infarction. Researchers and clinicians around the world have been engaged to regenerate the damaged human heart for a long time. Stem cell therapy is an exciting newer therapy to treat cardiovascular diseases. Various types of stem cells have been used to revive the damaged myocardium after myocardial infarction, and they have overall demonstrated safety and moderate efficacy. The specific mechanisms by which these cells help in improving cardiac function are still not completely known. There is growing evidence that intracoronary bone marrow cell transplantation in patients with myocardial infarction beneficially affects the remodeling of the damaged myocardium. Our systematic review article aims to assess the effects and the future of stem cell therapy in patients with myocardial Infarction. We searched articles in PubMed, ScienceDirect, and Google Scholar. Thirty-one studies that included 2171 patients in total were analyzed. Most of these studies showed stem cell therapy is safe and well tolerated in patients, and modest improvements are seen in left ventricular functions with no major adverse effects. However, some studies showed no positive and clinically significant outcomes. So, more high-quality studies on a larger scale are required to support and confirm its efficacy in remodeling damaged myocardium after myocardial infarction. We should also perform studies to determine the timing of cell delivery that is best suited for stem cell therapy.
Collapse
Affiliation(s)
- Rinky A Botleroo
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Renu Bhandari
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Manipal College of Medical Sciences, Pokhara, NPL
| | - Rowan Ahmed
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Roaa Kareem
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mallika Gyawali
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nanditha Venkatesan
- Internal Medicine, All India Institute of Medical Sciences, Raipur, IND.,Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Opemipo D Ogeyingbo
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Saint James School of Medicine, Park Ridge, USA.,Public Health, Walden University, Minneapolis, USA
| | - Abeer O Elshaikh
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
15
|
Li J, Hu S, Zhu D, Huang K, Mei X, López de Juan Abad B, Cheng K. All Roads Lead to Rome (the Heart): Cell Retention and Outcomes From Various Delivery Routes of Cell Therapy Products to the Heart. J Am Heart Assoc 2021; 10:e020402. [PMID: 33821664 PMCID: PMC8174178 DOI: 10.1161/jaha.120.020402] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the past decades, numerous preclinical studies and several clinical trials have evidenced the feasibility of cell transplantation in treating heart diseases. Over the years, different delivery routes of cell therapy have emerged and broadened the width of the field. However, a common hurdle is shared by all current delivery routes: low cell retention. A myriad of studies confirm that cell retention plays a crucial role in the success of cell-mediated cardiac repair. It is important for any delivery route to maintain donor cells in the recipient heart for enough time to not only proliferate by themselves, but also to send paracrine signals to surrounding damaged heart cells and repair them. In this review, we first undertake an in-depth study of primary theories of cell loss, including low efficiency in cell injection, "washout" effects, and cell death, and then organize the literature from the past decade that focuses on cell transplantation to the heart using various cell delivery routes, including intracoronary injection, systemic intravenous injection, retrograde coronary venous injection, and intramyocardial injection. In addition to a recapitulation of these approaches, we also clearly evaluate their strengths and weaknesses. Furthermore, we conduct comparative research on the cell retention rate and functional outcomes of these delivery routes. Finally, we extend our discussion to state-of-the-art bioengineering techniques that enhance cell retention, as well as alternative delivery routes, such as intrapericardial delivery. A combination of these novel strategies and more accurate assessment methods will help to address the hurdle of low cell retention and boost the efficacy of cell transplantation to the heart.
Collapse
Affiliation(s)
- Junlang Li
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Shiqi Hu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Dashuai Zhu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Huang
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Xuan Mei
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| |
Collapse
|
16
|
Carresi C, Scicchitano M, Scarano F, Macrì R, Bosco F, Nucera S, Ruga S, Zito MC, Mollace R, Guarnieri L, Coppoletta AR, Gliozzi M, Musolino V, Maiuolo J, Palma E, Mollace V. The Potential Properties of Natural Compounds in Cardiac Stem Cell Activation: Their Role in Myocardial Regeneration. Nutrients 2021; 13:275. [PMID: 33477916 PMCID: PMC7833367 DOI: 10.3390/nu13010275] [Citation(s) in RCA: 12] [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: 12/19/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs), which include congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, and many other cardiac disorders, cause about 30% of deaths globally; representing one of the main health problems worldwide. Among CVDs, ischemic heart diseases (IHDs) are one of the major causes of morbidity and mortality in the world. The onset of IHDs is essentially due to an unbalance between the metabolic demands of the myocardium and its supply of oxygen and nutrients, coupled with a low regenerative capacity of the heart, which leads to great cardiomyocyte (CM) loss; promoting heart failure (HF) and myocardial infarction (MI). To date, the first strategy recommended to avoid IHDs is prevention in order to reduce the underlying risk factors. In the management of IHDs, traditional therapeutic options are widely used to improve symptoms, attenuate adverse cardiac remodeling, and reduce early mortality rate. However, there are no available treatments that aim to improve cardiac performance by replacing the irreversible damaged cardiomyocytes (CMs). Currently, heart transplantation is the only treatment being carried out for irreversibly damaged CMs. Hence, the discovery of new therapeutic options seems to be necessary. Interestingly, recent experimental evidence suggests that regenerative stem cell medicine could be a useful therapeutic approach to counteract cardiac damage and promote tissue regeneration. To this end, researchers are tasked with answering one main question: how can myocardial regeneration be stimulated? In this regard, natural compounds from plant extracts seem to play a particularly promising role. The present review will summarize the recent advances in our knowledge of stem cell therapy in the management of CVDs; focusing on the main properties and potential mechanisms of natural compounds in stimulating and activating stem cells for myocardial regeneration.
Collapse
Affiliation(s)
- Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Federica Scarano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Roberta Macrì
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Francesca Bosco
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Saverio Nucera
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Stefano Ruga
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Rocco Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
| | - Ernesto Palma
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy; (F.S.); (R.M.); (F.B.); (S.N.); (S.R.); (M.C.Z.); (R.M.); (L.G.); (A.R.C.); (M.G.); (V.M.); (J.M.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88100 Catanzaro, Italy
| |
Collapse
|
17
|
Evolution of Stem Cells in Cardio-Regenerative Therapy. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Riaud M, Martinez MC, Montero-Menei CN. Scaffolds and Extracellular Vesicles as a Promising Approach for Cardiac Regeneration after Myocardial Infarction. Pharmaceutics 2020; 12:E1195. [PMID: 33317141 PMCID: PMC7763019 DOI: 10.3390/pharmaceutics12121195] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022] Open
Abstract
Clinical studies have demonstrated the regenerative potential of stem cells for cardiac repair over the past decades, but their widespread use is limited by the poor tissue integration and survival obtained. Natural or synthetic hydrogels or microcarriers, used as cell carriers, contribute to resolving, in part, the problems encountered by providing mechanical support for the cells allowing cell retention, survival and tissue integration. Moreover, hydrogels alone also possess mechanical protective properties for the ischemic heart. The combined effect of growth factors with cells and an appropriate scaffold allow a therapeutic effect on myocardial repair. Despite this, the effects obtained with cell therapy remain limited and seem to be equivalent to the effects obtained with extracellular vesicles, key actors in intercellular communication. Extracellular vesicles have cardioprotective effects which, when combined proangiogenic properties with antiapoptotic and anti-inflammatory actions, make it possible to act on all the damages caused by ischemia. The evolution of biomaterial engineering allows us to envisage their association with new major players in cardiac therapy, extracellular vesicles, in order to limit undesirable effects and to envisage a transfer to the clinic. This new therapeutic approach could be associated with the release of growth factors to potentialized the beneficial effect obtained.
Collapse
Affiliation(s)
- Melody Riaud
- SOPAM, U1063, INSERM, UNIV Angers, SFR ICAT, F-49800 Angers, France;
- CRCINA, UMR 1232, INSERM, Université de Nantes, Université d’Angers, F-49933 Angers, France
| | | | | |
Collapse
|
19
|
Domae K, Miyagawa S, Toda K, Sawa Y. New treatment strategy for severe heart failure: combination of ventricular assist device and regenerative therapy. J Artif Organs 2020; 24:1-5. [PMID: 32564158 DOI: 10.1007/s10047-020-01185-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/10/2020] [Indexed: 11/28/2022]
Abstract
Heart transplantation and ventricular assist device for the patients with end-stage heart failure are limited by availability and durability due to limited donor or device-related complication. Thus, complementation or a new alternative is needed for the treatment of severe heart failure. Based on the results of basic experiments, we applied skeletal myoblast cell sheet transplantation in a clinical setting using cell-sheet methods with temperature-responsive dish for the treatment of heart failure patient from 2007. After confirming the safety of this treatment, we started a clinical trial of myoblast cell sheet transplantation as sole therapy. According to these results, in 2015, myoblast cell sheet transplantation with ischemic cardiomyopathy was approved by the Japanese government and now this treatment was covered by Japanese health insurance. Here we report our approach and future perspective of cardiac regenerative therapy using this new treatment method for severe heart failure including new strategy incorporating regenerative therapy in the conventional treatment of heart failure including VAD and heart transplantation.
Collapse
Affiliation(s)
- Keitaro Domae
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Koichi Toda
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
20
|
Turner D, Rieger AC, Balkan W, Hare JM. Clinical-based Cell Therapies for Heart Disease-Current and Future State. Rambam Maimonides Med J 2020; 11:RMMJ.10401. [PMID: 32374254 PMCID: PMC7202446 DOI: 10.5041/rmmj.10401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients have an ongoing unmet need for effective therapies that reverse the cellular and functional damage associated with heart damage and disease. The discovery that ~1%-2% of adult cardiomyocytes turn over per year provided the impetus for treatments that stimulate endogenous repair mechanisms that augment this rate. Preclinical and clinical studies provide evidence that cell-based therapy meets these therapeutic criteria. Recent and ongoing studies are focused on determining which cell type(s) works best for specific patient population(s) and the mechanism(s) by which these cells promote repair. Here we review clinical and preclinical stem cell studies and anticipate future directions of regenerative medicine for heart disease.
Collapse
Affiliation(s)
- Darren Turner
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Angela C. Rieger
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
21
|
Analyzing Impetus of Regenerative Cellular Therapeutics in Myocardial Infarction. J Clin Med 2020; 9:jcm9051277. [PMID: 32354170 PMCID: PMC7287592 DOI: 10.3390/jcm9051277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Both vasculature and myocardium in the heart are excessively damaged following myocardial infarction (MI), hence therapeutic strategies for treating MI hearts should concurrently aim for true cardiac repair by introducing new cardiomyocytes to replace lost or injured ones. Of them, mesenchymal stem cells (MSCs) have long been considered a promising candidate for cell-based therapy due to their unspecialized, proliferative differentiation potential to specific cell lineage and, most importantly, their capacity of secreting beneficial paracrine factors which further promote neovascularization, angiogenesis, and cell survival. As a consequence, the differentiated MSCs could multiply and replace the damaged tissues to and turn into tissue- or organ-specific cells with specialized functions. These cells are also known to release potent anti-fibrotic factors including matrix metalloproteinases, which inhibit the proliferation of cardiac fibroblasts, thereby attenuating fibrosis. To achieve the highest possible therapeutic efficacy of stem cells, the other interventions, including hydrogels, electrical stimulations, or platelet-derived biomaterials, have been supplemented, which have resulted in a narrow to broad range of outcomes. Therefore, this article comprehensively analyzed the progress made in stem cells and combinatorial therapies to rescue infarcted myocardium.
Collapse
|
22
|
Yang D, O’Brien CG, Ikeda G, Traverse JH, Taylor DA, Henry TD, Bolli R, Yang PC. Meta-analysis of short- and long-term efficacy of mononuclear cell transplantation in patients with myocardial infarction. Am Heart J 2020; 220:155-175. [PMID: 31821904 PMCID: PMC7173405 DOI: 10.1016/j.ahj.2019.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/04/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Mononuclear cells (MNCs) have been tested in clinical trials across multiple cardiovascular pathologies with mixed results. Major adverse cardiac events (MACE) and markers of cardiovascular capacity have been particularly challenging to interpret because of small size. This meta-analysis is aimed to assess the efficacy of MNC therapy in randomized clinical trials to identify the markers of efficiency that could influence future trial design. METHODS PubMed, Embase, Cochrane library, and ClinicalTrials.gov were searched from inception through November 8, 2018. Changes in left ventricular ejection fraction (LVEF) and infarct size from baseline to follow-up were selected as primary outcomes. Changes in the left ventricular end-systolic volume, left ventricular end-diastolic volume, brain natriuretic peptide/N-terminal pro-B-type natriuretic peptide, 6-minute walk test, New York Heart Association class, and MACE incidences were considered secondary outcomes. RESULTS In short-term follow-up, patients treated with MNCs demonstrated a significant increase in absolute LVEF of 2.21% (95% CI 1.59-2.83; P < .001; I2 = 32%) and 6.01% (95% CI 4.45-7.57; P < .001; I2 = 0%) in acute myocardial infarction (AMI) and ischemic cardiomyopathy studies, respectively. This effect was sustained in long-term follow-up. MNC therapy significantly reduced left ventricular end-systolic volume; however, infarct size, 6-minute walk test, New York Heart Association class, and MACE rates were comparable. CONCLUSIONS MNC therapy may convey a modest but sustained increase in LVEF in ischemic cardiomyopathy patients, supporting further investigation. AMI trials, however, demonstrated minimal improvement in LVEF of unclear clinical significance, suggesting a limited role for MNC therapy in AMI.
Collapse
Affiliation(s)
- Dan Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China,Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Connor Galen O’Brien
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Gentaro Ikeda
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, MN55407, USA
| | - Doris A. Taylor
- Regenerative Medicine Research, Texas Heart Institute, PO Box 20345, Houston, TX 77225-0345 USA
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital Health Network, Cincinnati, OH45219, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, KY40202, USA
| | - Phillip C. Yang
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| |
Collapse
|
23
|
Sousa AR, Martins-Cruz C, Oliveira MB, Mano JF. One-Step Rapid Fabrication of Cell-Only Living Fibers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906305. [PMID: 31769556 DOI: 10.1002/adma.201906305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Cellular aggregates are used as relevant regenerative building blocks, tissue models, and cell delivery platforms. Biomaterial-free structures are often assembled either as 2D cell sheets or spherical microaggregates, both incompatible with free-form deposition, and dependent on challenging processes for macroscale 3D upscaling. The continuous and elongated nature of fiber-shaped materials enables their deposition in unrestricted multiple directions. Cellular fiber fabrication has often required exogenously provided support proteins and/or the use of biomaterial-based sacrificial templates. Here, the rapid (<24 h) assembly of fiberoids is reported: living centimeter-long scaffold-free fibers of cells produced in the absence of exogenous materials or supplements. Adipose-derived mesenchymal stem cell fiberoids can be easily modulated into complex multidimensional geometries and show tissue-invasive properties while keeping the secretion of trophic factors. Proangiogenic properties studied on a chick chorioallantoic membrane in an ovo model are observed for heterotypic fiberoids containing endothelial cells. These micro-to-macrotissues may find application as morphogenic therapeutic and tissue-mimetic building blocks, with the ability to integrate 3D and 4D full biological materials.
Collapse
Affiliation(s)
- Ana Rita Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Cláudia Martins-Cruz
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| |
Collapse
|
24
|
Traverse JH, Swingen CM, Henry TD, Fox J, Wang YL, Chavez IJ, Lips DL, Lesser JR, Pedersen WR, Burke NM, Pai A, Lindberg JL, Garberich RF. NHLBI-Sponsored Randomized Trial of Postconditioning During Primary Percutaneous Coronary Intervention for ST-Elevation Myocardial Infarction. Circ Res 2019; 124:769-778. [PMID: 30602360 DOI: 10.1161/circresaha.118.314060] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
RATIONALE Postconditioning at the time of primary percutaneous coronary intervention (PCI) for ST-segment-elevation myocardial infarction may reduce infarct size and improve myocardial salvage. However, clinical trials have shown inconsistent benefit. OBJECTIVE We performed the first National Heart, Lung, and Blood Institute-sponsored trial of postconditioning in the United States using strict enrollment criteria to optimize the early benefits of postconditioning and assess its long-term effects on left ventricular (LV) function. METHODS AND RESULTS We randomized 122 ST-segment-elevation myocardial infarction patients to postconditioning (4, 30 seconds PTCA [percutaneous transluminal coronary angioplasty] inflations/deflations)+PCI (n=65) versus routine PCI (n=57). All subjects had an occluded major epicardial artery (thrombolysis in myocardial infarction=0) with ischemic times between 1 and 6 hours with no evidence of preinfarction angina or collateral blood flow. Cardiac magnetic resonance imaging measured at 2 days post-PCI showed no difference between the postconditioning group and control in regards to infarct size (22.5±14.5 versus 24.0±18.5 g), myocardial salvage index (30.3±15.6% versus 31.5±23.6%), or mean LV ejection fraction. Magnetic resonance imaging at 12 months showed a significant recovery of LV ejection fraction in both groups (61.0±11.4% and 61.4±9.1%; P<0.01). Subjects randomized to postconditioning experienced more favorable remodeling over 1 year (LV end-diastolic volume =157±34 to 150±38 mL) compared with the control group (157±40 to 165±45 mL; P<0.03) and reduced microvascular obstruction ( P=0.05) on baseline magnetic resonance imaging and significantly less adverse LV remodeling compared with control subjects with microvascular obstruction ( P<0.05). No significant adverse events were associated with the postconditioning protocol and all patients but one (hemorrhagic stroke) survived through 1 year of follow-up. CONCLUSIONS We found no early benefit of postconditioning on infarct size, myocardial salvage index, and LV function compared with routine PCI. However, postconditioning was associated with improved LV remodeling at 1 year of follow-up, especially in subjects with microvascular obstruction. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT01324453.
Collapse
Affiliation(s)
- Jay H Traverse
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.).,Cardiovascular Division, The University of Minnesota School of Medicine, Minneapolis (J.H.T., C.M.S.)
| | - Cory M Swingen
- Cardiovascular Division, The University of Minnesota School of Medicine, Minneapolis (J.H.T., C.M.S.)
| | - Timothy D Henry
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Jane Fox
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Yale L Wang
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Ivan J Chavez
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Daniel L Lips
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - John R Lesser
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Wesley R Pedersen
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Nicholas M Burke
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Akila Pai
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Jana L Lindberg
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| | - Ross F Garberich
- From the Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T., T.D.H., J.F., Y.L.W., I.J.C., D.L.L., J.R.L., W.R.P., N.M.B., A.P., J.L.L., R.F.G.)
| |
Collapse
|
25
|
Abstract
The investment of nearly 2 decades of clinical investigation into cardiac cell therapy has yet to change cardiovascular practice. Recent insights into the mechanism of cardiac regeneration help explain these results and provide important context in which we can develop next-generation therapies. Non-contractile cells such as bone marrow or adult heart derivatives neither engraft long-term nor induce new muscle formation. Correspondingly, these cells offer little functional benefit to infarct patients. In contrast, preclinical data indicate that transplantation of bona fide cardiomyocytes derived from pluripotent stem cells induces direct remuscularization. This new myocardium beats synchronously with the host heart and induces substantial contractile benefits in macaque monkeys, suggesting that regeneration of contractile myocardium is required to fully recover function. Through a review of the preclinical and clinical trials of cardiac cell therapy, distinguishing the primary mechanism of benefit as either contractile or non-contractile helps appreciate the barriers to cardiac repair and establishes a rational path to optimizing therapeutic benefit.
Collapse
Affiliation(s)
- Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington
- Center for Cardiovascular Biology, University of Washington
- Department of Medicine/Cardiology, University of Washington
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington
- Center for Cardiovascular Biology, University of Washington
- Department of Medicine/Cardiology, University of Washington
- Department of Pathology, University of Washington
- Department of Bioengineering, University of Washington
| |
Collapse
|
26
|
Xu J, Xiong Y, Li Q, Hu M, Huang P, Xu J, Tian X, Jin C, Liu J, Qian L, Yang Y. Optimization of Timing and Times for Administration of Atorvastatin-Pretreated Mesenchymal Stem Cells in a Preclinical Model of Acute Myocardial Infarction. Stem Cells Transl Med 2019; 8:1068-1083. [PMID: 31245934 PMCID: PMC6766601 DOI: 10.1002/sctm.19-0013] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/25/2019] [Indexed: 12/14/2022] Open
Abstract
Our previous studies showed that the combination of atorvastatin (ATV) and single injection of ATV-pretreated mesenchymal stem cells (MSCs) (ATV -MSCs) at 1 week post-acute myocardial infarction (AMI) promoted MSC recruitment and survival. This study aimed to investigate whether the combinatorial therapy of intensive ATV with multiple injections of ATV -MSCs has greater efficacy at different stages to better define the optimal strategy for MSC therapy in AMI. In order to determine the optimal time window for MSC treatment, we first assessed stromal cell-derived factor-1 (SDF-1) dynamic expression and inflammation. Next, we compared MSC recruitment and differentiation, cardiac function, infarct size, and angiogenesis among animal groups with single, dual, and triple injections of ATV -MSCs at early (Early1, Early2, Early3), mid-term (Mid1, Mid2, Mid3), and late (Late1, Late2, Late3) stages. Compared with AMI control, intensive ATV significantly augmented SDF-1 expression 1.5∼2.6-fold in peri-infarcted region with inhibited inflammation. ATV -MSCs implantation with ATV administration further enhanced MSC recruitment rate by 3.9%∼24.0%, improved left ventricular ejection fraction (LVEF) by 2.0%∼16.2%, and reduced infarct size in all groups 6 weeks post-AMI with most prominent improvement in mid groups and still effective in late groups. Mechanistically, ATV -MSCs remarkably suppressed inflammation and apoptosis while increasing angiogenesis. Furthermore, triple injections of ATV -MSCs were much more effective than single administration during early and mid-term stages of AMI with the best effects in Mid3 group. We conclude that the optimal strategy is multiple injections of ATV -MSCs combined with intensive ATV administration at mid-term stage of AMI. The translational potential of this strategy is clinically promising. Stem Cells Translational Medicine 2019;8:1068-1083.
Collapse
Affiliation(s)
- Jun Xu
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
- McAllister Heart Institute, University of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
| | - Yu‐Yan Xiong
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Qing Li
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Meng‐Jin Hu
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Pei‐Sen Huang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
- McAllister Heart Institute, University of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
| | - Jun‐Yan Xu
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Xia‐Qiu Tian
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Chen Jin
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Jian‐Dong Liu
- McAllister Heart Institute, University of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
| | - Li Qian
- McAllister Heart Institute, University of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUnited States
| | - Yue‐Jin Yang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| |
Collapse
|
27
|
Banerjee MN, Bolli R, Hare JM. Clinical Studies of Cell Therapy in Cardiovascular Medicine: Recent Developments and Future Directions. Circ Res 2019; 123:266-287. [PMID: 29976692 DOI: 10.1161/circresaha.118.311217] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the rising prevalence of cardiovascular disease worldwide and the limited therapeutic options for severe heart failure, novel technologies that harness the regenerative capacity of the heart are sorely needed. The therapeutic use of stem cells has the potential to reverse myocardial injury and improve cardiac function, in contrast to most current medical therapies that only mitigate heart failure symptoms. Nearly 2 decades and >200 trials for cardiovascular disease have revealed that most cell types are safe; however, their efficacy remains controversial, limiting the transition of this therapy from investigation to practice. Lessons learned from these initial studies are driving the design of new clinical trials; higher fidelity of cell isolation techniques, standardization of conditions, more consistent use of state of the art measurement techniques, and assessment of multiple end points to garner insights into the efficacy of stem cells. Translation to clinical trials has almost outpaced our mechanistic understanding, and individual patient factors likely play a large role in stem cell efficacy. Therefore, careful analysis of dosing, delivery methods, and the ideal patient populations is necessary to translate cell therapy from research to practice. We are at a pivotal stage in the field in which information from many relatively small clinical trials must guide carefully executed efficacy trials. Larger efficacy trials are being launched to answer questions about older, first-generation stem cell therapeutics, while novel, second-generation products are being introduced into the clinical realm. This review critically examines the current state of clinical research on cell-based therapies for cardiovascular disease, highlighting the controversies in the field, improvements in clinical trial design, and the application of exciting new cell products.
Collapse
Affiliation(s)
- Monisha N Banerjee
- From the Interdisciplinary Stem Cell Institute (M.N.B., J.M.H.).,Department of Surgery (M.N.B)
| | - Roberto Bolli
- University of Miami Miller School of Medicine, FL; and Institute of Molecular Cardiology, University of Louisville, KY (R.B.)
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (M.N.B., J.M.H.) .,Department of Medicine (J.M.H.)
| |
Collapse
|
28
|
Affiliation(s)
- Jeffrey M Tyler
- From the Cedars-Sinai Smidt Heart Institute, Los Angeles, CA (J.M.T., T.D.H.)
| | | | - Timothy D Henry
- From the Cedars-Sinai Smidt Heart Institute, Los Angeles, CA (J.M.T., T.D.H.)
| |
Collapse
|
29
|
Sant'Anna RT, Eibel B, Markoski MM, Rodrigues CG, de Salles FB, Giusti II, Nesralla IA, Nardi NB, Kalil RAK. Gene therapy for refractory angina and cell therapy for heart failure: experience of a Brazilian research group. Gene Ther 2019; 27:40-50. [PMID: 31278371 DOI: 10.1038/s41434-019-0087-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
Abstract
Cell therapy has shown impressive effects in experimental cardiomyopathy models. To a lesser extent, gene therapy has also been studied. In both cases, translation to clinical therapy has been disappointing. This paper is intended to describe the experience and achievements of a multicenter working group located in Porto Alegre, southern Brazil, in experimental and translational research projects for cell-based and gene therapy methods in the treatment of dilated and ischemic cardiomyopathies. The results of preclinical and clinical studies showed that bone marrow mononuclear stem cells indeed have an effect in improving myocardial perfusion and contractile function, but the overall results are poorly translated to the clinical level. Gene therapy studies with direct myocardial injections of naked VEGF 165 plasmid showed improvement in myocardial perfusion and function in animal models. A randomized clinical trial found that this method is safe and improved myocardial perfusion, but the benefits disappeared after 1 year. An animal experiment associating VEGF 165 with angiopoietin was undertaken in mini pigs to extend the durability of that therapy. In conclusion, our efforts to better understand the mechanisms and functions of gene and cell-based therapies in cardiology resulted in significant findings and propose a future look at cell-free therapeutic approaches.
Collapse
Affiliation(s)
- Roberto Tofani Sant'Anna
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil
| | - Bruna Eibel
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil
| | - Melissa Medeiros Markoski
- Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite 245, 90050-170, Porto Alegre, RS, Brazil
| | - Clarissa Garcia Rodrigues
- Global Research and Innovation Network - GRINN, Rua Doutor João Colin 1285, Sala 03, 89204-001, Joinville, SC, Brazil
| | - Felipe Borsu de Salles
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil
| | - Imarilde Inês Giusti
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil
| | - Ivo Abrahão Nesralla
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil
| | - Nance Beyer Nardi
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil.,Universidade Luterana do Brasil, Av Farroupilha, 8001, 92425-900, Canoas, RS, Brazil
| | - Renato Abdala Karam Kalil
- Instituto de Cardiologia do Rio Grande do Sul, Av Princesa Isabel 395, 90620-001, Porto Alegre, RS, Brazil.
| |
Collapse
|
30
|
Williams R. Circulation Research "In This Issue" Anthology. Circ Res 2019; 124:e123-e148. [PMID: 31170049 DOI: 10.1161/res.0000000000000275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
31
|
Panahi M, Vadgama N, Kuganesan M, Ng FS, Sattler S. Immunopharmacology of Post-Myocardial Infarction and Heart Failure Medications. J Clin Med 2018; 7:E403. [PMID: 30384415 PMCID: PMC6262592 DOI: 10.3390/jcm7110403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 10/23/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
The immune system responds to acute tissue damage after myocardial infarction (MI) and orchestrates healing and recovery of the heart. However, excessive inflammation may lead to additional tissue damage and fibrosis and exacerbate subsequent functional impairment, leading to heart failure. The appreciation of the immune system as a crucial factor after MI has led to a surge of clinical trials investigating the potential benefits of immunomodulatory agents previously used in hyper-inflammatory conditions, such as autoimmune disease. While the major goal of routine post-MI pharmacotherapy is to support heart function by ensuring appropriate blood pressure and cardiac output to meet the demands of the body, several drug classes also affect a range of immunological pathways and modulate the post-MI immune response, which is crucial to take into account when designing future immunomodulatory trials. This review outlines how routine post-MI pharmacotherapy affects the immune response and may thus influence post-MI outcomes and development towards heart failure. Current key drug classes are discussed, including platelet inhibitors, statins, β-blockers, and renin⁻angiotensin⁻aldosterone inhibitors.
Collapse
Affiliation(s)
- Mona Panahi
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| | - Nimai Vadgama
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| | - Mathun Kuganesan
- University College London Medical School, University College London, London WC1E 6BT, UK.
| | - Fu Siong Ng
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
32
|
Panahi M, Papanikolaou A, Torabi A, Zhang JG, Khan H, Vazir A, Hasham MG, Cleland JGF, Rosenthal NA, Harding SE, Sattler S. Immunomodulatory interventions in myocardial infarction and heart failure: a systematic review of clinical trials and meta-analysis of IL-1 inhibition. Cardiovasc Res 2018; 114:1445-1461. [PMID: 30010800 PMCID: PMC6106100 DOI: 10.1093/cvr/cvy145] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/26/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Following a myocardial infarction (MI), the immune system helps to repair ischaemic damage and restore tissue integrity, but excessive inflammation has been implicated in adverse cardiac remodelling and development towards heart failure (HF). Pre-clinical studies suggest that timely resolution of inflammation may help prevent HF development and progression. Therapeutic attempts to prevent excessive post-MI inflammation in patients have included pharmacological interventions ranging from broad immunosuppression to immunomodulatory approaches targeting specific cell types or factors with the aim to maintain beneficial aspects of the early post-MI immune response. These include the blockade of early initiators of inflammation including reactive oxygen species and complement, inhibition of mast cell degranulation and leucocyte infiltration, blockade of inflammatory cytokines, and inhibition of adaptive B and T-lymphocytes. Herein, we provide a systematic review on post-MI immunomodulation trials and a meta-analysis of studies targeting the inflammatory cytokine Interleukin-1. Despite an enormous effort into a significant number of clinical trials on a variety of targets, a striking heterogeneity in study population, timing and type of treatment, and highly variable endpoints limits the possibility for meaningful meta-analyses. To conclude, we highlight critical considerations for future studies including (i) the therapeutic window of opportunity, (ii) immunological effects of routine post-MI medication, (iii) stratification of the highly diverse post-MI patient population, (iv) the potential benefits of combining immunomodulatory with regenerative therapies, and at last (v) the potential side effects of immunotherapies.
Collapse
Affiliation(s)
- Mona Panahi
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, UK
| | - Angelos Papanikolaou
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, UK
| | - Azam Torabi
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, Sydney Street, London, UK
| | - Ji-Gang Zhang
- The Jackson Laboratory, 600 Main Street, Bar Harbor, USA
| | - Habib Khan
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, Sydney Street, London, UK
| | - Ali Vazir
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, Sydney Street, London, UK
| | | | - John G F Cleland
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, UK
- Royal Brompton Hospital, Royal Brompton and Harefield NHS Foundation Trust, Sydney Street, London, UK
| | - Nadia A Rosenthal
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, UK
- The Jackson Laboratory, 600 Main Street, Bar Harbor, USA
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, UK
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London, UK
| |
Collapse
|
33
|
Bolli R, Hare JM, March KL, Pepine CJ, Willerson JT, Perin EC, Yang PC, Henry TD, Traverse JH, Mitrani RD, Khan A, Hernandez-Schulman I, Taylor DA, DiFede DL, Lima JAC, Chugh A, Loughran J, Vojvodic RW, Sayre SL, Bettencourt J, Cohen M, Moyé L, Ebert RF, Simari RD. Rationale and Design of the CONCERT-HF Trial (Combination of Mesenchymal and c-kit + Cardiac Stem Cells As Regenerative Therapy for Heart Failure). Circ Res 2018; 122:1703-1715. [PMID: 29703749 DOI: 10.1161/circresaha.118.312978] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/16/2018] [Accepted: 04/25/2018] [Indexed: 12/15/2022]
Abstract
RATIONALE Autologous bone marrow mesenchymal stem cells (MSCs) and c-kit+ cardiac progenitor cells (CPCs) are 2 promising cell types being evaluated for patients with heart failure (HF) secondary to ischemic cardiomyopathy. No information is available in humans about the relative efficacy of MSCs and CPCs and whether their combination is more efficacious than either cell type alone. OBJECTIVE CONCERT-HF (Combination of Mesenchymal and c-kit+ Cardiac Stem Cells As Regenerative Therapy for Heart Failure) is a phase II trial aimed at elucidating these issues by assessing the feasibility, safety, and efficacy of transendocardial administration of autologous MSCs and CPCs, alone and in combination, in patients with HF caused by chronic ischemic cardiomyopathy (coronary artery disease and old myocardial infarction). METHODS AND RESULTS Using a randomized, double-blinded, placebo-controlled, multicenter, multitreatment, and adaptive design, CONCERT-HF examines whether administration of MSCs alone, CPCs alone, or MSCs+CPCs in this population alleviates left ventricular remodeling and dysfunction, reduces scar size, improves quality of life, or augments functional capacity. The 4-arm design enables comparisons of MSCs alone with CPCs alone and with their combination. CONCERT-HF consists of 162 patients, 18 in a safety lead-in phase (stage 1) and 144 in the main trial (stage 2). Stage 1 is complete, and stage 2 is currently randomizing patients from 7 centers across the United States. CONCLUSIONS CONCERT-HF will provide important insights into the potential therapeutic utility of MSCs and CPCs, given alone and in combination, for patients with HF secondary to ischemic cardiomyopathy. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT02501811.
Collapse
Affiliation(s)
- Roberto Bolli
- From the Division of Cardiovascular Medicine, University of Louisville, KY (R.B., J.L.)
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H., A.K., R.D.M., I.H.-S.)
| | - Keith L March
- Division of Cardiovascular Medicine, UFHealth at University of Florida, Gainesville (K.L.M., C.J.P.)
| | - Carl J Pepine
- Division of Cardiovascular Medicine, UFHealth at University of Florida, Gainesville (K.L.M., C.J.P.)
| | - James T Willerson
- Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., D.A.T.)
| | - Emerson C Perin
- Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., D.A.T.)
| | - Phillip C Yang
- Cardiovascular Medicine, Stanford University School of Medicine, CA (P.C.Y.)
| | - Timothy D Henry
- Division of Cardiology, Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.)
| | - Jay H Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN (J.H.T.)
| | - Raul D Mitrani
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H., A.K., R.D.M., I.H.-S.)
| | - Aisha Khan
- From the Division of Cardiovascular Medicine, University of Louisville, KY (R.B., J.L.)
| | - Ivonne Hernandez-Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL (J.M.H., A.K., R.D.M., I.H.-S.)
| | - Doris A Taylor
- Texas Heart Institute, CHI St. Luke's Health, Houston (J.T.W., E.C.P., D.A.T.)
| | | | - João A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD (J.A.C.L.)
| | - Atul Chugh
- Franciscan Saint Francis Health, Indianapolis, IN (A.C.)
| | - John Loughran
- From the Division of Cardiovascular Medicine, University of Louisville, KY (R.B., J.L.)
| | - Rachel W Vojvodic
- Coordinating Center for Clinical Trials, UT Health School of Public Health, Houston, TX (R.W.V., S.L.S., J.B., M.C., L.M.)
| | - Shelly L Sayre
- Coordinating Center for Clinical Trials, UT Health School of Public Health, Houston, TX (R.W.V., S.L.S., J.B., M.C., L.M.)
| | - Judy Bettencourt
- Coordinating Center for Clinical Trials, UT Health School of Public Health, Houston, TX (R.W.V., S.L.S., J.B., M.C., L.M.)
| | - Michelle Cohen
- Coordinating Center for Clinical Trials, UT Health School of Public Health, Houston, TX (R.W.V., S.L.S., J.B., M.C., L.M.)
| | - Lem Moyé
- Coordinating Center for Clinical Trials, UT Health School of Public Health, Houston, TX (R.W.V., S.L.S., J.B., M.C., L.M.)
| | - Ray F Ebert
- NIH, National Heart, Lung, and Blood Institute, Division of Cardiovascular Sciences, Bethesda, MD (R.F.E.)
| | | | | |
Collapse
|
34
|
Abstract
Despite considerable advances in medicine, cardiovascular disease is still rising, with ischemic heart disease being the leading cause of death and disability worldwide. Thus extensive efforts are continuing to establish effective therapeutic modalities that would improve both quality of life and survival in this patient population. Novel therapies are being investigated not only to protect the myocardium against ischemia-reperfusion injury but also to regenerate the heart. Stem cell therapy, such as potential use of human mesenchymal stem cells and induced pluripotent stem cells and their exosomes, will make it possible not only to address molecular mechanisms of cardiac conditioning, but also to develop new therapies for ischemic heart disease. Despite the studies and progress made over the last 15 years on the use of stem cell therapy for cardiovascular disease, the efforts are still in their infancy. Even though the expectations have been high, the findings indicate that most of the clinical trials generally have been small and the results inconclusive. Because of many negative findings, there is certain pessimism that cardiac cell therapy is likely to yield any meaningful results over the next decade or so. Similar to other new technologies, early failures are not unusual and they may be followed by impressive success. Nevertheless, there has been considerable attention to safety by the clinical investigators because the adverse events of stem cell therapy have been impressively rare. In summary, although regenerative biology might not help the cardiovascular patient in the near term, it is destined to do so over the next several decades.
Collapse
Affiliation(s)
- Maia Terashvili
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI
| | - Zeljko J Bosnjak
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
35
|
Madigan M, Atoui R. Therapeutic Use of Stem Cells for Myocardial Infarction. Bioengineering (Basel) 2018; 5:bioengineering5020028. [PMID: 29642402 PMCID: PMC6027340 DOI: 10.3390/bioengineering5020028] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022] Open
Abstract
Myocardial infarction is a leading cause of morbidity and mortality worldwide. Although medical and surgical treatments can significantly improve patient outcomes, no treatment currently available is able to generate new contractile tissue or reverse ischemic myocardium. Driven by the recent/novel understanding that regenerative processes do exist in the myocardium—tissue previously thought not to possess regenerative properties—the use of stem cells has emerged as a promising therapeutic approach with high expectations. The literature describes the use of cells from various sources, categorizing them as either embryonic, induced pluripotent, or adult/tissue stem cells (mesenchymal, hematopoietic, skeletal myoblasts, cardiac stem cells). Many publications show the successful use of these cells to regenerate damaged myocardium in both animal and human models; however, more studies are needed to directly compare cells of various origins in efforts to draw conclusions on the ideal source. Although numerous challenges exist in this developing area of research and clinical practice, prospects are encouraging. The following aims to provide a concise review outlining the different types of stem cells used in patients after myocardial infarction.
Collapse
Affiliation(s)
- Mariah Madigan
- Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada.
| | - Rony Atoui
- Health Sciences North, Sudbury, ON P3E 5J1, Canada.
| |
Collapse
|
36
|
Abstract
Recently in Cell, Mohamed et al. (2018) report a cell-cycle regulator gene cocktail identified from young cardiomyocytes that enables mouse, rat, and human cardiomyocyte proliferation and promotes heart regeneration after infarction, defying the non-dividing nature of adult mammalian cardiomyocytes and implying a new way to treat or prevent heart failure.
Collapse
|
37
|
Mittal R, Jhaveri VM, McMurry HS, Kay SIS, Sutherland KJ, Nicole L, Mittal J, Jayant RD. Recent treatment modalities for cardiovascular diseases with a focus on stem cells, aptamers, exosomes and nanomedicine. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:831-840. [PMID: 29447002 DOI: 10.1080/21691401.2018.1436555] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Due to the significant impact of CVD on humans, there is a need to develop novel treatment modalities tailored to major classes of cardiac diseases including hypertension, coronary artery disease, cardiomyopathies, arrhythmias, valvular disease and inflammatory diseases. In this article, we discuss recent advancements regarding development of therapeutic strategies based on stem cells, aptamers, exosomes, drug-eluting and dissolvable stents, immunotherapy and nanomedicine for the treatment of CVD. We summarize current research and clinical advances in cardiovascular therapeutics, with a focus on therapies that move beyond current oral- or sublingual-based regimens. This review article provides insight into current research and future treatment strategies that hold a great relevance for future clinical practice in pursuit of improving quality of life of patients suffering from CVD.
Collapse
Affiliation(s)
- Rahul Mittal
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Vasanti M Jhaveri
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Hannah S McMurry
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Sae-In Samantha Kay
- b Dr. Kiran C. Patel College of Osteopathic Medicine , Nova Southeastern University , Fort Lauderdale , FL , USA
| | - Kyle J Sutherland
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Lin Nicole
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Jeenu Mittal
- a Department of Otolaryngology , University of Miami Miller School of Medicine , Miami , FL , USA
| | - Rahul Dev Jayant
- c Department of Immunology , Center for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University , Miami , FL , USA
| |
Collapse
|