1
|
Li J, Gong G, Zhang Y, Zheng Y, He Y, Chen M, He X, Zheng X, Gong X, Liu L, Zhou K, Zhao Z, Iv CWS, Hua Y, Li Y, Guo J. Polyphenol-Nanoengineered Monocyte Biohybrids for Targeted Cardiac Repair and Immunomodulation. Adv Healthc Mater 2025; 14:e2403595. [PMID: 39526529 DOI: 10.1002/adhm.202403595] [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: 09/20/2024] [Revised: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Myocardial infarction is one of the leading cause of cardiovascular death worldwide. Invasive interventional procedures and medications are applied to attenuate the attacks associated with ischemic heart disease by reestablishing blood flow and restoring oxygen supply. However, the overactivation of inflammatory responses and unsatisfactory drug delivery efficiency in the infarcted regions prohibit functional improvement. Here, a nanoengineered monocyte (MO)-based biohybrid system, referred to as CTAs @MOs, for the heart-targeted delivery of combinational therapeutic agents (CTAs) containing anti-inflammatory IL-10 and cardiomyogenic miR-19a to overcome the limitation of malperfusion within the infarcted myocardium through a polyphenol-mediated interfacial assembly, is reported. Systemic administration of CTAs@MOs bypasses extensive thoracotomy and intramyocardial administration risks, leading to infarcted heart-specific accumulation and sustained release of therapeutic agents, enabling immunomodulation of the proinflammatory microenvironment and promoting cardiomyocyte proliferation in sequence. Moreover, CTAs@MOs, which serve as a cellular biohybrid-based therapy, significantly improve cardiac function as evidenced by enhanced ejection fractions, increased fractional shortening, and diminished infarct sizes. This polyphenol nanoengineered biohybrid system represents a general and potent platform for the efficient treatment of cardiovascular disorders.
Collapse
Affiliation(s)
- Jiawen Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Guidong Gong
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yue Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanjiang Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunxiang He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Mei Chen
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xianglian He
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xiaolan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue Gong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - C Wyatt Shields Iv
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Colorado, 80303, USA
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatric Dentistry, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Junling Guo
- BMI Center for Biomass Materials and Nanointerfaces, National Engineering Laboratory for Clean Technology of Leather Manufacture, Ministry of Education Key Laboratory of Leather Chemistry and Engineering, College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Department of Chemical and Biological Engineering, The University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
2
|
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] [Grants] [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
|
3
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
4
|
Ye IB, Hines GL. Therapeutic Angiogenesis and Cardiovascular Disease: A Review. Cardiol Rev 2024:00045415-990000000-00284. [PMID: 38814076 DOI: 10.1097/crd.0000000000000729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
After the success of novel angiogenesis inhibitors in cancer treatment, angiogenesis promotors for the treatment of peripheral vascular disease and coronary artery disease became the target of significant research. Promising results in animal models led to numerous randomized control trials that failed to translate into meaningful clinical results. The goal of this review is to describe the history of investigation into therapeutic angiogenesis for cardiovascular disease and discuss the lessons learned and future directions.
Collapse
Affiliation(s)
- Ivan B Ye
- From the Division of Vascular Surgery, NYU Langone Hospital-Long Island, Mineola, NY
| | | |
Collapse
|
5
|
Olatunji G, Kokori E, Yusuf I, Ayanleke E, Damilare O, Afolabi S, Adetunji B, Mohammed S, Akinmoju O, Aboderin G, Aderinto N. Stem cell-based therapies for heart failure management: a narrative review of current evidence and future perspectives. Heart Fail Rev 2024; 29:573-598. [PMID: 37733137 DOI: 10.1007/s10741-023-10351-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 09/22/2023]
Abstract
Heart failure (HF) is a prevalent and debilitating global cardiovascular condition affecting around 64 million individuals, placing significant strain on healthcare systems and diminishing patients' quality of life. The escalating prevalence of HF underscores the urgent need for innovative therapeutic approaches that target the root causes and aim to restore normal cardiac function. Stem cell-based therapies have emerged as promising candidates, representing a fundamental departure from conventional treatments focused primarily on symptom management. This review explores the evolving landscape of stem cell-based therapies for HF management. It delves into the mechanisms of action, clinical evidence from both positive and negative outcomes, ethical considerations, and regulatory challenges. Key findings include the potential for improved cardiac function, enhanced quality of life, and long-term benefits associated with stem cell therapies. However, adverse events and patient vulnerabilities necessitate stringent safety assessments. Future directions in stem cell-based HF therapies include enhancing efficacy and safety through optimized stem cell types, delivery techniques, dosing strategies, and long-term safety assessments. Personalized medicine, combining therapies, addressing ethical and regulatory challenges, and expanding access while reducing costs are crucial aspects of the evolving landscape.
Collapse
Affiliation(s)
- Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Ismaila Yusuf
- Department of Medicine and Surgery, Obafemi Awolowo University, Osun, Nigeria
| | - Emmanuel Ayanleke
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Olakanmi Damilare
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Samson Afolabi
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Busayo Adetunji
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Saad Mohammed
- Al-Kindy College of Medicine, University of Baghdad, Baghdad, Iraq
| | | | - Gbolahan Aboderin
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria
| | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University Teaching Hospital, Ogbomoso, Nigeria.
| |
Collapse
|
6
|
Carvalho AB, Kasai-Brunswick TH, Campos de Carvalho AC. Advanced cell and gene therapies in cardiology. EBioMedicine 2024; 103:105125. [PMID: 38640834 PMCID: PMC11052923 DOI: 10.1016/j.ebiom.2024.105125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024] Open
Abstract
We review the evidence for the presence of stem/progenitor cells in the heart and the preclinical and clinical data using diverse cell types for the therapy of cardiac diseases. We highlight the failure of adult stem/progenitor cells to ameliorate heart function in most cardiac diseases, with the possible exception of refractory angina. The use of pluripotent stem cell-derived cardiomyocytes is analysed as a viable alternative therapeutic option but still needs further research at preclinical and clinical stages. We also discuss the use of direct reprogramming of cardiac fibroblasts into cardiomyocytes and the use of extracellular vesicles as therapeutic agents in ischemic and non-ischemic cardiac diseases. Finally, gene therapies and genome editing for the treatment of hereditary cardiac diseases, ablation of genes responsible for atherosclerotic disease, or modulation of gene expression in the heart are discussed.
Collapse
Affiliation(s)
- Adriana Bastos Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Universidade Federal do RIo de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tais Hanae Kasai-Brunswick
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Universidade Federal do RIo de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonio Carlos Campos de Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Universidade Federal do RIo de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
7
|
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
|
8
|
Wang Y, Chen L, Wang L, Pei G, Cheng H, Zhang Q, Wang S, Hu D, He Y, He C, Fu C, Wei Q. Pulsed Electromagnetic Fields Combined With Adipose-Derived Stem Cells Protect Ischemic Myocardium by Regulating miR-20a-5p/E2F1/p73 Signaling. Stem Cells 2023; 41:724-737. [PMID: 37207995 DOI: 10.1093/stmcls/sxad037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/03/2023] [Indexed: 05/21/2023]
Abstract
Myocardial infarction (MI) is a serious threat to human health. Although monotherapy with pulsed electromagnetic fields (PEMFs) or adipose-derived stem cells (ADSCs) has been reported to have positive effect on the treatment of MI, a satisfactory outcome has not yet been achieved. In recent years, combination therapy has attracted widespread interest. Herein, we explored the synergistic therapeutic effect of combination therapy with PEMFs and ADSCs on MI and found that the combination of PEMFs and ADSCs effectively reduced infarct size, inhibited cardiomyocyte apoptosis and protected the cardiac function in mice with MI. In addition, bioinformatics analysis and RT-qPCR showed that the combination therapy could affect apoptosis by regulating the expression of miR-20a-5p. A dual-luciferase reporter gene assay also confirmed that the miR-20a-5p could target E2F transcription factor 1 (E2F1) and inhibit cardiomyocyte apoptosis by regulating the E2F1/p73 signaling pathway. Therefore, our study systematically demonstrated the effectiveness of combination therapy on the inhibition of cardiomyocyte apoptosis by regulating the miR-20a-5p/E2F1/p73 signaling pathway in mice with MI. Thus, our study underscored the effectiveness of the combination of PEMFs and ADSCs and identified miR-20a-5p as a promising therapeutic target for the treatment of MI in the future.
Collapse
Affiliation(s)
- Yang Wang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Li Chen
- Department of Rehabilitation Medicine, The Fifth Affiliated People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Lu Wang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Gaiqin Pei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Hongxin Cheng
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Qing Zhang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Shiqi Wang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Danrong Hu
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Yong He
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengqi He
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Quan Wei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People's Republic of China
| |
Collapse
|
9
|
Gallet R, Su JB, Corboz D, Chiaroni PM, Bizé A, Dai J, Panel M, Boucher P, Pallot G, Brehat J, Sambin L, Thery G, Mouri N, de Pommereau A, Denormandie P, Germain S, Lacampagne A, Teiger E, Marbán E, Ghaleh B. Three-vessel coronary infusion of cardiosphere-derived cells for the treatment of heart failure with preserved ejection fraction in a pre-clinical pig model. Basic Res Cardiol 2023; 118:26. [PMID: 37400630 DOI: 10.1007/s00395-023-00995-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/05/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major public health concern. Its outcome is poor and, as of today, barely any treatments have been able to decrease its morbidity or mortality. Cardiosphere-derived cells (CDCs) are heart cell products with anti-fibrotic, anti-inflammatory and angiogenic properties. Here, we tested the efficacy of CDCs in improving left ventricular (LV) structure and function in pigs with HFpEF. Fourteen chronically instrumented pigs received continuous angiotensin II infusion for 5 weeks. LV function was investigated through hemodynamic measurements and echocardiography at baseline, after 3 weeks of angiotensin II infusion before three-vessel intra-coronary CDC (n = 6) or placebo (n = 8) administration and 2 weeks after treatment (i.e., at completion of the protocol). As expected, arterial pressure was significantly and similarly increased in both groups. This was accompanied by LV hypertrophy that was not affected by CDCs. LV systolic function remained similarly preserved during the whole protocol in both groups. In contrast, LV diastolic function was impaired (increases in Tau, LV end-diastolic pressure as well as E/A, E/E'septal and E/E'lateral ratios) but CDC treatment significantly improved all of these parameters. The beneficial effect of CDCs on LV diastolic function was not explained by reduced LV hypertrophy or increased arteriolar density; however, interstitial fibrosis was markedly reduced. Three-vessel intra-coronary administration of CDCs improves LV diastolic function and reduces LV fibrosis in this hypertensive model of HFpEF.
Collapse
Affiliation(s)
- Romain Gallet
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Service de Cardiologie, Créteil, France
| | - Jin-Bo Su
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Daphné Corboz
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Paul-Matthieu Chiaroni
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Service de Cardiologie, Créteil, France
| | - Alain Bizé
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Jianping Dai
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Mathieu Panel
- PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Pierre Boucher
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Gaëtan Pallot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Juliette Brehat
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Lucien Sambin
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Guillaume Thery
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Nadir Mouri
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de biochimie-pharmacologie-biologie moléculaire-génétique médicale, Créteil, France
| | - Aurélien de Pommereau
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Pierre Denormandie
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Alain Lacampagne
- PhyMedExp, Université de Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Emmanuel Teiger
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Service de Cardiologie, Créteil, France
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Bijan Ghaleh
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.
| |
Collapse
|
10
|
Leancă SA, Afrăsânie I, Crișu D, Matei IT, Duca ȘT, Costache AD, Onofrei V, Tudorancea I, Mitu O, Bădescu MC, Șerban LI, Costache II. Cardiac Reverse Remodeling in Ischemic Heart Disease with Novel Therapies for Heart Failure with Reduced Ejection Fraction. Life (Basel) 2023; 13:1000. [PMID: 37109529 PMCID: PMC10143569 DOI: 10.3390/life13041000] [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: 03/07/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the improvements in the treatment of coronary artery disease (CAD) and acute myocardial infarction (MI) over the past 20 years, ischemic heart disease (IHD) continues to be the most common cause of heart failure (HF). In clinical trials, over 70% of patients diagnosed with HF had IHD as the underlying cause. Furthermore, IHD predicts a worse outcome for patients with HF, leading to a substantial increase in late morbidity, mortality, and healthcare costs. In recent years, new pharmacological therapies have emerged for the treatment of HF, such as sodium-glucose cotransporter-2 inhibitors, angiotensin receptor-neprilysin inhibitors, selective cardiac myosin activators, and oral soluble guanylate cyclase stimulators, demonstrating clear or potential benefits in patients with HF with reduced ejection fraction. Interventional strategies such as cardiac resynchronization therapy, cardiac contractility modulation, or baroreflex activation therapy might provide additional therapeutic benefits by improving symptoms and promoting reverse remodeling. Furthermore, cardiac regenerative therapies such as stem cell transplantation could become a new therapeutic resource in the management of HF. By analyzing the existing data from the literature, this review aims to evaluate the impact of new HF therapies in patients with IHD in order to gain further insight into the best form of therapeutic management for this large proportion of HF patients.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Afrăsânie
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Crișu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ștefania Teodora Duca
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Viviana Onofrei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ionuţ Tudorancea
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ovidiu Mitu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Iuliana Costache
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
11
|
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
|
12
|
Xu CM, Karbasiafshar C, Brinck Teixeira R, Ahsan N, Blume Corssac G, Sellke FW, Abid MR. Proteomic Assessment of Hypoxia-Pre-Conditioned Human Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles Demonstrates Promise in the Treatment of Cardiovascular Disease. Int J Mol Sci 2023; 24:1674. [PMID: 36675188 PMCID: PMC9866304 DOI: 10.3390/ijms24021674] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Human bone marrow mesenchymal stem cell derived-extracellular vesicles (HBMSC-EV) are known for their regenerative and anti-inflammatory effects in animal models of myocardial ischemia. However, it is not known whether the efficacy of the EVs can be modulated by pre-conditioning of HBMSC by exposing them to either starvation or hypoxia prior to EV collection. HBMSC-EVs were isolated following normoxia starvation (NS), normoxia non-starvation (NNS), hypoxia starvation (HS), or hypoxia non-starvation (HNS) pre-conditioning. The HBMSC-EVs were characterized by nanoparticle tracking analysis, electron microscopy, Western blot, and proteomic analysis. Comparative proteomic profiling revealed that starvation pre-conditioning led to a smaller variety of proteins expressed, with the associated lesser effect of normoxia versus hypoxia pre-conditioning. In the absence of starvation, normoxia and hypoxia pre-conditioning led to disparate HBMSC-EV proteomic profiles. HNS HBMSC-EV was found to have the greatest variety of proteins overall, with 74 unique proteins, the greatest number of redox proteins, and pathway analysis suggestive of improved angiogenic properties. Future HBMSC-EV studies in the treatment of cardiovascular disease may achieve the most therapeutic benefits from hypoxia non-starved pre-conditioned HBMSC. This study was limited by the lack of functional and animal models of cardiovascular disease and transcriptomic studies.
Collapse
Affiliation(s)
- Cynthia M. Xu
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | | | - Rayane Brinck Teixeira
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Nagib Ahsan
- Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK 73019, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| | - Giana Blume Corssac
- Cardiovascular Physiology Laboratory, Basic Health Sciences Institute, UFRGS, Porto Alegre, RS, Brazil
| | - Frank W. Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - M. Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
13
|
Okamura A, Yoshioka Y, Saito Y, Ochiya T. Can Extracellular Vesicles as Drug Delivery Systems Be a Game Changer in Cardiac Disease? Pharm Res 2022; 40:889-908. [PMID: 36577860 PMCID: PMC10126064 DOI: 10.1007/s11095-022-03463-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022]
Abstract
Cardiac diseases such as myocardial infarction and heart failure have been the leading cause of death worldwide for more than 20 years, and new treatments continue to be investigated. Heart transplantation, a curative treatment for severe cardiac dysfunction, is available to only a small number of patients due to the rarity of donors and high costs. Cardiac regenerative medicine using embryonic stem cells and induced pluripotent stem cells is expected to be a new alternative to heart transplantation, but it has problems such as induction of immune response, tumor formation, and low survival rate of transplanted cells. On the other hand, there has been a focus on cell-free therapy using extracellular vesicles (EVs) due to their high biocompatibility and target specificity. Exosomes, one type of EV, play a role in the molecular transport system in vivo and can be considered a drug delivery system (DDS) innate to all living things. Exosomes contain nucleic acids and proteins, which are transported from secretory cells to recipient cells. Molecules in exosomes are encapsulated in a lipid bilayer, which allows them to exist stably in body fluids without being affected by nuclease degradation enzymes. Therefore, the therapeutic use of exosomes as DDSs has been widely explored and is being used in clinical trials and other clinical settings. This review summarizes the current topics of EVs as DDSs in cardiac disease.
Collapse
Affiliation(s)
- Akihiko Okamura
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.,Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
14
|
Pu X, Zhu P, Zhou X, He Y, Wu H, Du L, Gong H, Sun X, Chen T, Zhu J, Xu Q, Zhang H. CD34 + cell atlas of main organs implicates its impact on fibrosis. Cell Mol Life Sci 2022; 79:576. [PMID: 36315271 PMCID: PMC11803001 DOI: 10.1007/s00018-022-04606-6] [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: 05/06/2022] [Revised: 09/22/2022] [Accepted: 10/11/2022] [Indexed: 11/03/2022]
Abstract
RATIONALE CD34+ cells are believed being progenitors that may be used to treat cardiovascular disease. However, the exact identity and the role of CD34+ cells in physiological and pathological conditions remain unclear. METHODS We performed single-cell RNA sequencing analysis to provide a cell atlas of normal tissue/organ and pathological conditions. Furthermore, a genetic lineage tracing mouse model was used to investigate the role of CD34+ cells in angiogenesis and organ fibrosis. RESULTS Single-cell RNA sequencing analysis revealed a heterogeneous population of CD34+ cells in both physiological and pathological conditions. Using a genetic lineage tracing mouse model, we showed that CD34+ cells not only acquired endothelial cell fate involved in angiogenesis, but also, CD34+ cells expressing Pi16 may transform into myofibroblast and thus participate in organ fibrosis. CONCLUSION A heterogeneous CD34+ cells serve as a contributor not only to endothelial regeneration but also a wound healing response that may provide therapeutic insights into fibrosis.
Collapse
Affiliation(s)
- Xiangyuan Pu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Pengwei Zhu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Xuhao Zhou
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Yangyan He
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Wu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Hui Gong
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Xiaotong Sun
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| | - Jianhua Zhu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China.
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, Hangzhou, China.
| | - Hongkun Zhang
- Department of Vascular Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
15
|
Wang Y, Xue Y, Guo HD. Intervention effects of traditional Chinese medicine on stem cell therapy of myocardial infarction. Front Pharmacol 2022; 13:1013740. [PMID: 36330092 PMCID: PMC9622800 DOI: 10.3389/fphar.2022.1013740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases are the leading cause of global mortality, in which myocardial infarction accounts for 46% of total deaths. Although good progress has been achieved in medication and interventional techniques, a proven method to repair the damaged myocardium has not yet been determined. Stem cell therapy for damaged myocardial repair has evolved into a promising treatment for ischemic heart disease. However, low retention and poor survival of the injected stem cells are the major obstacles to achieving the intended therapeutic effects. Chinese botanical and other natural drug substances are a rich source of effective treatment for various diseases. As such, numerous studies have revealed the role of Chinese medicine in stem cell therapy for myocardial infarction treatment, including promoting proliferation, survival, migration, angiogenesis, and differentiation of stem cells. Here, we discuss the potential and limitations of stem cell therapy, as well as the regulatory mechanism of Chinese medicines underlying stem cell therapy. We focus on the evidence from pre-clinical trials and clinical practices, and based on traditional Chinese medicine theories, we further summarize the mechanisms of Chinese medicine treatment in stem cell therapy by the commonly used prescriptions. Despite the pre-clinical evidence showing that traditional Chinese medicine is helpful in stem cell therapy, there are still some limitations of traditional Chinese medicine therapy. We also systematically assess the detailed experimental design and reliability of included pharmacological research in our review. Strictly controlled animal models with multi-perspective pharmacokinetic profiles and high-grade clinical evidence with multi-disciplinary efforts are highly demanded in the future.
Collapse
Affiliation(s)
- Yu Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuezhen Xue
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hai-dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
16
|
Assessment of Myocardial Diastolic Dysfunction as a Result of Myocardial Infarction and Extracellular Matrix Regulation Disorders in the Context of Mesenchymal Stem Cell Therapy. J Clin Med 2022; 11:jcm11185430. [PMID: 36143077 PMCID: PMC9502668 DOI: 10.3390/jcm11185430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
The decline in cardiac contractility due to damage or loss of cardiomyocytes is intensified by changes in the extracellular matrix leading to heart remodeling. An excessive matrix response in the ischemic cardiomyopathy may contribute to the elevated fibrotic compartment and diastolic dysfunction. Fibroproliferation is a defense response aimed at quickly closing the damaged area and maintaining tissue integrity. Balance in this process is of paramount importance, as the reduced post-infarction response causes scar thinning and more pronounced left ventricular remodeling, while excessive fibrosis leads to impairment of heart function. Under normal conditions, migration of progenitor cells to the lesion site occurs. These cells have the potential to differentiate into myocytes in vitro, but the changed micro-environment in the heart after infarction does not allow such differentiation. Stem cell transplantation affects the extracellular matrix remodeling and thus may facilitate the improvement of left ventricular function. Studies show that mesenchymal stem cell therapy after infarct reduces fibrosis. However, the authors did not specify whether they meant the reduction of scarring as a result of regeneration or changes in the matrix. Research is also necessary to rule out long-term negative effects of post-acute infarct stem cell therapy.
Collapse
|
17
|
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]
|
18
|
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: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [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
|
19
|
Zhang X, Wu Y, Cheng Q, Bai L, Huang S, Gao J. Extracellular Vesicles in Cardiovascular Diseases: Diagnosis and Therapy. Front Cell Dev Biol 2022; 10:875376. [PMID: 35721498 PMCID: PMC9198246 DOI: 10.3389/fcell.2022.875376] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality. Therapy of CVDs is still a great challenge since many advanced therapies have been developed. Multiple cell types produce nano-sized extracellular vesicles (EVs), including cardiovascular system-related cells and stem cells. Compelling evidence reveals that EVs are associated with the pathophysiological processes of CVDs. Recently researches focus on the clinical transformation in EVs-based diagnosis, prognosis, therapies, and drug delivery systems. In this review, we firstly discuss the current knowledge about the biophysical properties and biological components of EVs. Secondly, we will focus on the functions of EVs on CVDs, and outline the latest advances of EVs as prognostic and diagnostic biomarkers, and therapeutic agents. Finally, we will introduce the specific application of EVs as a novel drug delivery system and its application in CVDs therapy. Specific attention will be paid to summarize the perspectives, challenges, and applications on EVs’ clinical and industrial transformation.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
- *Correspondence: Xiaojing Zhang, ; Jun Gao,
| | - Yuping Wu
- Department of Scientific Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Qifa Cheng
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Liyang Bai
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Shuqiang Huang
- Department of Clinical Medicine, The Sixth Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Jun Gao
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
- *Correspondence: Xiaojing Zhang, ; Jun Gao,
| |
Collapse
|
20
|
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: 3.3] [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
|
21
|
Frantz S, Hundertmark MJ, Schulz-Menger J, Bengel FM, Bauersachs J. Left ventricular remodelling post-myocardial infarction: pathophysiology, imaging, and novel therapies. Eur Heart J 2022; 43:2549-2561. [PMID: 35511857 PMCID: PMC9336586 DOI: 10.1093/eurheartj/ehac223] [Citation(s) in RCA: 249] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 04/13/2022] [Indexed: 12/11/2022] Open
Abstract
Most patients survive acute myocardial infarction (MI). Yet this encouraging development has certain drawbacks: heart failure (HF) prevalence is increasing and patients affected tend to have more comorbidities worsening economic strain on healthcare systems and impeding effective medical management. The heart’s pathological changes in structure and/or function, termed myocardial remodelling, significantly impact on patient outcomes. Risk factors like diabetes, chronic obstructive pulmonary disease, female sex, and others distinctly shape disease progression on the ‘road to HF’. Despite the availability of HF drugs that interact with general pathways involved in myocardial remodelling, targeted drugs remain absent, and patient risk stratification is poor. Hence, in this review, we highlight the pathophysiological basis, current diagnostic methods and available treatments for cardiac remodelling following MI. We further aim to provide a roadmap for developing improved risk stratification and novel medical and interventional therapies.
Collapse
Affiliation(s)
- Stefan Frantz
- Department of Internal Medicine I, Universitätsklinikum Würzburg, University Hospital Würzburg, University of Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Moritz Jens Hundertmark
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jeanette Schulz-Menger
- Department of Cardiology and Nephrology, Experimental and Clinical Research Center, a Joint Cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine and HELIOS Hospital Berlin Buch, Berlin, Germany
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Miloradovic D, Miloradovic D, Ljujic B, Jankovic MG. Optimal Delivery Route of Mesenchymal Stem Cells for Cardiac Repair: The Path to Good Clinical Practice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022:83-100. [PMID: 35389200 DOI: 10.1007/5584_2022_709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Research has shown that mesenchymal stem cells (MSCs) could be a promising therapy for treating progressive heart disease. However, translation into clinics efficiently and successfully has proven to be much more complicated. Many questions remain for optimizing treatment. Application method influences destiny of MSCs and afterwards impacts results of procedure, yet there is no general agreement about most suitable method of MSC delivery in the clinical setting. Herein, we explain principle of most-frequent MSCs delivery techniques in cardiology. This chapter summarizes crucial translational obstacles of clinical employment of MSCs for cardiac repair when analysed trough a prism of latest research centred on different techniques of MSCs application.
Collapse
Affiliation(s)
- Dragica Miloradovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Miloradovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Biljana Ljujic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia
| | - Marina Gazdic Jankovic
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, Kragujevac, Serbia.
| |
Collapse
|
23
|
Dutzmann J, Knöpp K, Kefalianakis Z, Daniel JM, Gufler H, Wohlgemuth W, Kahles F, Sedding DG. Effect of intermittent fasting after ST-elevation myocardial infarction on left ventricular function: study protocol of a pilot randomised controlled trial (INTERFAST-MI). BMJ Open 2022; 12:e050067. [PMID: 35393305 PMCID: PMC8990699 DOI: 10.1136/bmjopen-2021-050067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Preclinical studies consistently show robust disease-modifying effects of intermittent fasting in animal models of cardiovascular disease. However, the impact of intermittent fasting on cardiovascular endpoints after myocardial infarction has not been investigated in a clinical trial so far. METHODS AND ANALYSIS The INTERmittent FASTing after Myocardial Infarction (INTERFAST-MI) trial is a monocentric prospective randomised controlled non-confirmatory pilot study including 48 patients with ST-segment elevation myocardial infarction. They will be randomised in a 1:1 ratio to either intermittent fasting (daily time-restricted eating; consuming food for not more than 8 hours/day, fasting for at least 16 hours/day) or to a control group without a particular diet. The follow-up time is 6 months. The prespecified primary outcome is change in left ventricular systolic function at 4 weeks from baseline to estimate effect size required to establishing sample size and power calculation for a future full-scale trial. Secondary outcomes include protocol adherence, recruitment, major adverse cardiac events, revascularisation, changes in left ventricular systolic function at 3 and 6 months, patient weight, blood pressure, and serum markers of inflammation and cardiovascular disease. Enrolment began on 1 November 2020 and is expected to conclude in December 2021. ETHICS AND DISSEMINATION The trial has received ethics approval from the Medical Ethics Committee of the Martin-Luther-University Halle-Wittenberg. Results of the study will be submitted for publication in a peer-reviewed journal and presented at scientific conferences. TRIAL REGISTRATION NUMBER DRKS00021784.
Collapse
Affiliation(s)
- Jochen Dutzmann
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Kai Knöpp
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Zoe Kefalianakis
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Jan-Marcus Daniel
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| | - Hubert Gufler
- Department of Radiology, University Hospital Halle, Halle, Germany
| | | | - Florian Kahles
- Department of Internal Medicine I, University Hospital Aachen, Aachen, Germany
| | - Daniel G Sedding
- Department of Internal Medicine III, University Hospital Halle, Halle, Germany
| |
Collapse
|
24
|
Lee CS, Kim J, Cho HJ, Kim HS. Cardiovascular Regeneration via Stem Cells and Direct Reprogramming: A Review. Korean Circ J 2022; 52:341-353. [PMID: 35502566 PMCID: PMC9064703 DOI: 10.4070/kcj.2022.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Despite recent advancements in treatment strategies, cardiovascular disease such as heart failure remains a significant source of global mortality. Stem cell technology and cellular reprogramming are rapidly growing fields that will continue to prove useful in cardiac regenerative therapeutics. This review provides information on the role of human pluripotent stem cells (hPSCs) in cardiac regeneration and discusses the practical applications of hPSC-derived cardiomyocytes (CMCs). Moreover, we discuss the practical applications of hPSC-derived CMCs while outlining the relevance of directly-reprogrammed CMCs in regenerative medicine. This review critically summarizes the most recent advances in the field will help to guide future research in this developing area. Cardiovascular disease (CVD) is the leading causes of morbidity and death globally. In particular, a heart failure remains a major problem that contributes to global mortality. Considerable advancements have been made in conventional pharmacological therapies and coronary intervention surgery for cardiac disorder treatment. However, more than 15% of patients continuously progress to end-stage heart failure and eventually require heart transplantation. Over the past year, numerous numbers of protocols to generate cardiomyocytes (CMCs) from human pluripotent stem cells (hPSCs) have been developed and applied in clinical settings. Number of studies have described the therapeutic effects of hPSCs in animal models and revealed the underlying repair mechanisms of cardiac regeneration. In addition, biomedical engineering technologies have improved the therapeutic potential of hPSC-derived CMCs in vivo. Recently substantial progress has been made in driving the direct differentiation of somatic cells into mature CMCs, wherein an intermediate cellular reprogramming stage can be bypassed. This review provides information on the role of hPSCs in cardiac regeneration and discusses the practical applications of hPSC-derived CMCs; furthermore, it outlines the relevance of directly reprogrammed CMCs in regenerative medicine.
Collapse
Affiliation(s)
- Choon-Soo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
25
|
Xu S, Qiu Y, Tao J. The challenges and optimization of cell-based therapy for cardiovascular disease. J Transl Int Med 2021; 9:234-238. [PMID: 35136722 PMCID: PMC8802397 DOI: 10.2478/jtim-2021-0017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
With the hope of achieving real cardiovascular repair, cell-based therapy raised as a promising strategy for the treatment of cardiovascular disease (CVD) in the past two decades. Various types of cells have been studied for their reparative potential for CVD in the ensuing years. Despite the exciting results from animal experiments, the outcome of clinical trials is unsatisfactory and the development of cell-based therapy for CVD has hit a plateau nowadays. Thus, it is important to summarize the obstacles we are facing in this field in order to explore possible solutions for optimizing cell-based therapy and achieving real clinical application.
Collapse
Affiliation(s)
- Shiyue Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou510080, Guangdong Province, China
- Department of Biomedical Engineering, Molecular Cardiology Program, School of Medicine and School of Engineering, University of Alabama at BirminghamBirminghamUnited States
| | - Yumin Qiu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou510080, Guangdong Province, China
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou510080, Guangdong Province, China
| |
Collapse
|
26
|
Pluripotent stem cell-derived mesenchymal stromal cells improve cardiac function and vascularity after myocardial infarction. Cytotherapy 2021; 23:1074-1084. [PMID: 34588150 DOI: 10.1016/j.jcyt.2021.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have been shown to improve cardiac function after injury and are the subject of ongoing clinical trials. In this study, the authors tested the cardiac regenerative potential of an induced pluripotent stem cell-derived MSC (iPSC-MSC) population (Cymerus MSCs) in a rat model of myocardial ischemia-reperfusion (I/R). Furthermore, the authors compared this efficacy with bone marrow-derived MSCs (BM-MSCs), which are the predominant cell type in clinical trials. METHODS Four days after myocardial I/R injury, rats were randomly assigned to (i) a Cymerus MSC group (n = 15), (ii) a BM-MSC group (n = 15) or (iii) a vehicle control group (n = 14). For cell-treated animals, a total of 5 × 106 cells were injected at three sites within the infarcted left ventricular (LV) wall. RESULTS One month after cell transplantation, Cymerus MSCs improved LV function (assessed by echocardiography) compared with vehicle and BM-MSCs. Interestingly, Cymerus MSCs enhanced angiogenesis without sustained engraftment or significant impact on infarct scar size. Suggesting safety, Cymerus MSCs had no effect on inducible tachycardia or the ventricular scar heterogeneity that provides a substrate for cardiac re-entrant circuits. CONCLUSIONS The authors here demonstrate that intra-myocardial administration of iPSC-MSCs (Cymerus MSCs) provide better therapeutic effects compared with conventional BM-MSCs in a rodent model of myocardial I/R. Because of its manufacturing scalability, iPSC-MSC therapy offers an exciting opportunity for an "off-the-shelf" stem cell therapy for cardiac repair.
Collapse
|
27
|
Das S, Nam H, Jang J. 3D bioprinting of stem cell-laden cardiac patch: A promising alternative for myocardial repair. APL Bioeng 2021; 5:031508. [PMID: 34368602 PMCID: PMC8318604 DOI: 10.1063/5.0030353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 06/01/2021] [Indexed: 12/18/2022] Open
Abstract
Stem cell-laden three-dimensional (3D) bioprinted cardiac patches offer an alternative and promising therapeutic and regenerative approach for ischemic cardiomyopathy by reversing scar formation and promoting myocardial regeneration. Numerous studies have reported using either multipotent or pluripotent stem cells or their combination for 3D bioprinting of a cardiac patch with the sole aim of restoring cardiac function by faithfully rejuvenating the cardiomyocytes and associated vasculatures that are lost to myocardial infarction. While many studies have demonstrated success in mimicking cardiomyocytes' behavior, improving cardiac function and providing new hope for regenerating heart post-myocardial infarction, some others have reported contradicting data in apparent ways. Nonetheless, all investigators in the field are speed racing toward determining a potential strategy to effectively treat losses due to myocardial infarction. This review discusses various types of candidate stem cells that possess cardiac regenerative potential, elucidating their applications and limitations. We also brief the challenges of and an update on the implementation of the state-of-the-art 3D bioprinting approach to fabricate cardiac patches and highlight different strategies to implement vascularization and augment cardiac functional properties with respect to electrophysiological similarities to native tissue.
Collapse
Affiliation(s)
- Sanskrita Das
- Department of Convergence IT Engineering, POSTECH, 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Hyoryung Nam
- Department of Convergence IT Engineering, POSTECH, 77 Cheongam-ro, Namgu, Pohang, Kyungbuk 37673, Republic of Korea
| | - Jinah Jang
- Author to whom correspondence should be addressed:
| |
Collapse
|
28
|
Herrmann JL. Commentary: Another iteration of cell-based therapy for acute ischemia-reperfusion injury, this time in the spine. JTCVS OPEN 2021; 7:41-42. [PMID: 36003756 PMCID: PMC9390552 DOI: 10.1016/j.xjon.2021.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Jeremy L. Herrmann
- Address for reprints: Jeremy L. Herrmann, MD, Indiana University School of Medicine, 545 Barnhill Dr, Emerson 215, Indianapolis, IN 46202.
| |
Collapse
|
29
|
Zaki MM, Lesha E, Said K, Kiaee K, Robinson-McCarthy L, George H, Hanna A, Appleton E, Liu S, Ng AHM, Khoshakhlagh P, Church GM. Cell therapy strategies for COVID-19: Current approaches and potential applications. SCIENCE ADVANCES 2021; 7:eabg5995. [PMID: 34380619 PMCID: PMC8357240 DOI: 10.1126/sciadv.abg5995] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/22/2021] [Indexed: 05/03/2023]
Abstract
Coronavirus disease 2019 (COVID-19) continues to burden society worldwide. Despite most patients having a mild course, severe presentations have limited treatment options. COVID-19 manifestations extend beyond the lungs and may affect the cardiovascular, nervous, and other organ systems. Current treatments are nonspecific and do not address potential long-term consequences such as pulmonary fibrosis, demyelination, and ischemic organ damage. Cell therapies offer great potential in treating severe COVID-19 presentations due to their customizability and regenerative function. This review summarizes COVID-19 pathogenesis, respective areas where cell therapies have potential, and the ongoing 89 cell therapy trials in COVID-19 as of 1 January 2021.
Collapse
Affiliation(s)
- Mark M Zaki
- GC Therapeutics Inc., Cambridge, MA 02139, USA
- Department of Neurosurgery, University of Michigan, 1500 E Medical Center Dr., Ann Arbor, MI 48109, USA
| | - Emal Lesha
- GC Therapeutics Inc., Cambridge, MA 02139, USA
- Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Khaled Said
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kiavash Kiaee
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | - Angy Hanna
- Department of Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Evan Appleton
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Songlei Liu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Alex H M Ng
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - Parastoo Khoshakhlagh
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| | - George M Church
- GC Therapeutics Inc., Cambridge, MA 02139, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA
| |
Collapse
|
30
|
Ostovaneh MR, Makkar RR, Ambale-Venkatesh B, Ascheim D, Chakravarty T, Henry TD, Kowalchuk G, Aguirre FV, Kereiakes DJ, Povsic TJ, Schatz R, Traverse JH, Pogoda J, Smith RD, Marbán L, Marbán E, Lima JAC. Effect of cardiosphere-derived cells on segmental myocardial function after myocardial infarction: ALLSTAR randomised clinical trial. Open Heart 2021; 8:e001614. [PMID: 34233913 PMCID: PMC8264869 DOI: 10.1136/openhrt-2021-001614] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Most cell therapy trials failed to show an improvement in global left ventricular (LV) function measures after myocardial infarction (MI). Myocardial segments are heterogeneously impacted by MI. Global LV function indices are not able to detect the small treatment effects on segmental myocardial function which may have prognostic implications for cardiac events. We aimed to test the efficacy of allogeneic cardiosphere-derived cells (CDCs) for improving regional myocardial function and contractility. METHODS In this exploratory analysis of a randomised clinical trial, 142 patients with post-MI with LVEF <45% and 15% or greater LV scar size were randomised in 2:1 ratio to receive intracoronary infusion of allogenic CDCs or placebo, respectively. Change in segmental myocardial circumferential strain (Ecc) by MRI from baseline to 6 months was compared between CDCs and placebo groups. RESULTS In total, 124 patients completed the 6-month follow-up (mean (SD) age 54.3 (10.8) and 108 (87.1%) men). Segmental Ecc improvement was significantly greater in patients receiving CDC (-0.5% (4.0)) compared with placebo (0.2% (3.7), p=0.05). The greatest benefit for improvement in segmental Ecc was observed in segments containing scar tissue (change in segmental Ecc of -0.7% (3.5) in patients receiving CDC vs 0.04% (3.7) in the placebo group, p=0.04). CONCLUSIONS In patients with post-MI LV dysfunction, CDC administration resulted in improved segmental myocardial function. Our findings highlight the importance of segmental myocardial function indices as an endpoint in future clinical trials of patients with post-MI. TRIAL REGISTRATION NUMBER NCT01458405.
Collapse
Affiliation(s)
- Mohammad R Ostovaneh
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medicine, Penn State Milton S Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Raj R Makkar
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angles, California, USA
| | | | | | - Tarun Chakravarty
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angles, California, USA
| | | | - Glen Kowalchuk
- Sanger Heart and Vascular Institute, Charlotte, North Carolina, USA
| | | | | | - Thomas J Povsic
- Duke Clinical Research Institute and Duke Medicine, Durham, North Carolina, USA
| | | | - Jay H Traverse
- Minneapolis Heart Institute Foundation, Minneapolis, Minnesota, USA
| | - Janice Pogoda
- Cipher Biostatistics and Reporting, Reno, Nevada, USA
| | | | - Linda Marbán
- Capricor Therapeutics Inc, Los Angles, California, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angles, California, USA
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
31
|
Therapies to prevent post-infarction remodelling: From repair to regeneration. Biomaterials 2021; 275:120906. [PMID: 34139506 DOI: 10.1016/j.biomaterials.2021.120906] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is the first cause of worldwide mortality, with an increasing incidence also reported in developing countries. Over the past decades, preclinical research and clinical trials continually tested the efficacy of cellular and acellular-based treatments. However, none of them resulted in a drug or device currently used in combination with either percutaneous coronary intervention or coronary artery bypass graft. Inflammatory, proliferation and remodelling phases follow the ischaemic event in the myocardial tissue. Only recently, single-cell sequencing analyses provided insights into the specific cell populations which determine the final fibrotic deposition in the affected region. In this review, ischaemia, inflammation, fibrosis, angiogenesis, cellular stress and fundamental cellular and molecular components are evaluated as therapeutic targets. Given the emerging evidence of biomaterial-based systems, the increasing use of injectable hydrogels/scaffolds and epicardial patches is reported both as acellular and cellularised/functionalised treatments. Since several variables influence the outcome of any experimented treatment, we return to the pathological basis with an unbiased view towards any specific process or cellular component. Thus, by evaluating the benefits and limitations of the approaches based on these targets, the reader can weigh the rationale of each of the strategies that reached the clinical trials stage. As recent studies focused on the relevance of the extracellular matrix in modulating ischaemic remodelling and enhancing myocardial regeneration, we aim to portray current trends in the field with this review. Finally, approaches towards feasible translational studies that are as yet unexplored are also suggested.
Collapse
|
32
|
Carbone RG, Monselise A, Bottino G, Negrini S, Puppo F. Stem cells therapy in acute myocardial infarction: a new era? Clin Exp Med 2021; 21:231-237. [PMID: 33484381 PMCID: PMC8053645 DOI: 10.1007/s10238-021-00682-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Stem cells transplantation after acute myocardial infarction (AMI) has been claimed to restore cardiac function. However, this therapy is still restricted to experimental studies and clinical trials. Early un-blinded studies suggested a benefit from stem cell therapy following AMI. More recent blinded randomized trials have produced mixed results and, notably, the last largest pan-European clinical trial showed the inconclusive results. Furthermore, mechanisms of potential benefit remain uncertain. This review analytically evaluates 34 blinded and un-blinded clinical trials comprising 3142 patients and is aimed to: (1) identify the pros and cons of stem cell therapy up to a 6-month follow-up after AMI comparing benefit or no effectiveness reported in clinical trials; (2) provide useful information for planning future clinical programs of cardiac stem cell therapy.
Collapse
Affiliation(s)
- R G Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | - G Bottino
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - S Negrini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - F Puppo
- Department of Internal Medicine, University of Genoa, Genoa, Italy.
| |
Collapse
|
33
|
Shin HS, Shin HH, Shudo Y. Current Status and Limitations of Myocardial Infarction Large Animal Models in Cardiovascular Translational Research. Front Bioeng Biotechnol 2021; 9:673683. [PMID: 33996785 PMCID: PMC8116580 DOI: 10.3389/fbioe.2021.673683] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/06/2021] [Indexed: 01/16/2023] Open
Abstract
Establishing an appropriate disease model that mimics the complexities of human cardiovascular disease is critical for evaluating the clinical efficacy and translation success. The multifaceted and complex nature of human ischemic heart disease is difficult to recapitulate in animal models. This difficulty is often compounded by the methodological biases introduced in animal studies. Considerable variations across animal species, modifications made in surgical procedures, and inadequate randomization, sample size calculation, blinding, and heterogeneity of animal models used often produce preclinical cardiovascular research that looks promising but is irreproducible and not translatable. Moreover, many published papers are not transparent enough for other investigators to verify the feasibility of the studies and the therapeutics' efficacy. Unfortunately, successful translation of these innovative therapies in such a closed and biased research is difficult. This review discusses some challenges in current preclinical myocardial infarction research, focusing on the following three major inhibitors for its successful translation: Inappropriate disease model, frequent modifications to surgical procedures, and insufficient reporting transparency.
Collapse
Affiliation(s)
- Hye Sook Shin
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Heather Hyeyoon Shin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Yasuhiro Shudo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
34
|
Xie DM, Chen Y, Liao Y, Lin W, Dai G, Lu DH, Zhu S, Yang K, Wu B, Chen Z, Peng C, Jiang MH. Cardiac Derived CD51-Positive Mesenchymal Stem Cells Enhance the Cardiac Repair Through SCF-Mediated Angiogenesis in Mice With Myocardial Infarction. Front Cell Dev Biol 2021; 9:642533. [PMID: 33968928 PMCID: PMC8098770 DOI: 10.3389/fcell.2021.642533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Many tissues contained resident mesenchymal stromal/stem cells (MSCs) that facilitated tissue hemostasis and repair. However, there is no typical marker to identify the resident cardiac MSCs. We aimed to determine if CD51 could be an optimal marker of cardiac MSCs and assess their therapeutic potential for mice with acute myocardial infarction (AMI). Methods: Cardiac-derived CD51+CD31–CD45–Ter119– cells (named CD51+cMSCs) were isolated from C57BL/6 mice(7-day-old) by flow cytometry. The CD51+cMSCs were characterized by proliferation capacity, multi-differentiation potential, and expression of typical MSC-related markers. Adult C57BL/6 mice (12-week-old) were utilized for an AMI model via permanently ligating the left anterior descending coronary artery. The therapeutic efficacy of CD51+cMSCs was estimated by echocardiography and pathological staining. To determine the underlying mechanism, lentiviruses were utilized to knock down gene (stem cell factor [SCF]) expression of CD51+cMSCs. Results: In this study, CD51 was expressed in the entire layers of the cardiac wall in mice, including endocardium, epicardium, and myocardium, and its expression was decreased with age. Importantly, the CD51+cMSCs possessed potent self-renewal potential and multi-lineage differentiation capacity in vitro and also expressed typical MSC-related surface proteins. Furthermore, CD51+cMSC transplantation significantly improved cardiac function and attenuated cardiac fibrosis through pro-angiogenesis activity after myocardial infarction in mice. Moreover, SCF secreted by CD51+cMSCs played an important role in angiogenesis both in vivo and in vitro. Conclusions: Collectively, CD51 is a novel marker of cardiac resident MSCs, and CD51+cMSC therapy enhances cardiac repair at least partly through SCF-mediated angiogenesis.
Collapse
Affiliation(s)
- Dong Mei Xie
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Yang Chen
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Wanwen Lin
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gang Dai
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Di Han Lu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuanghua Zhu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Yang
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bingyuan Wu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihong Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Chaoquan Peng
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Hua Jiang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Madeddu P. Cell therapy for the treatment of heart disease: Renovation work on the broken heart is still in progress. Free Radic Biol Med 2021; 164:206-222. [PMID: 33421587 DOI: 10.1016/j.freeradbiomed.2020.12.444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/26/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) continues to be the number one killer in the aging population. Heart failure (HF) is also an important cause of morbidity and mortality in patients with congenital heart disease (CHD). Novel therapeutic approaches that could restore stable heart function are much needed in both paediatric and adult patients. Regenerative medicine holds promises to provide definitive solutions for correction of congenital and acquired cardiac defects. In this review article, we recap some important aspects of cardiovascular cell therapy. First, we report quantifiable data regarding the scientific advancements in the field and how this has been translated into tangible outcomes according clinical studies and related meta-analyses. We then comment on emerging trends and technologies, such as the use of second-generation cell products, including pericyte-like vascular progenitors, and reprogramming of cells by different approaches including modulation of oxidative stress. The more affordable and feasible strategy of repurposing clinically available drugs to awaken the intrinsic healing potential of the heart will be discussed in the light of current social, financial, and ethical context. Cell therapy remains a work in progress field. Uncertainty in the ability of the experts and policy makers to solve urgent medical problems is growing in a world that is significantly influenced by them. This is particularly true in the field of regenerative medicine, due to great public expectations, polarization of leadership and funding, and insufficient translational vision. Cardiovascular regenerative medicine should be contextualized in a holistic program with defined priorities to allow a complete realization. Reshaping the notion of medical expertise is fundamental to fill the current gap in translation.
Collapse
Affiliation(s)
- Paolo Madeddu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, BS28HW, Bristol, United Kingdom.
| |
Collapse
|
36
|
Zhang R, Yu J, Zhang N, Li W, Wang J, Cai G, Chen Y, Yang Y, Liu Z. Bone marrow mesenchymal stem cells transfer in patients with ST-segment elevation myocardial infarction: single-blind, multicenter, randomized controlled trial. Stem Cell Res Ther 2021; 12:33. [PMID: 33413636 PMCID: PMC7791674 DOI: 10.1186/s13287-020-02096-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Our aim was to evaluate the efficacy and safety of intracoronary autologous bone marrow mesenchymal stem cell (BM-MSC) transplantation in patients with ST-segment elevation myocardial infarction (STEMI). METHODS In this randomized, single-blind, controlled trial, patients with STEMI (aged 39-76 years) were enrolled at 6 centers in Beijing (The People's Liberation Army Navy General Hospital, Beijing Armed Police General Hospital, Chinese People's Liberation Army General Hospital, Beijing Huaxin Hospital, Beijing Tongren Hospital, Beijing Chaoyang Hospital West Hospital). All patients underwent optimum medical treatment and percutaneous coronary intervention and were randomly assigned in a 1:1 ratio to BM-MSC group or control group. The primary endpoint was the change of myocardial viability at the 6th month's follow-up and left ventricular (LV) function at the 12th month's follow-up. The secondary endpoints were the incidence of cardiovascular event, total mortality, and adverse event during the 12 months' follow-up. The myocardial viability assessed by single-photon emission computed tomography (SPECT). The left ventricular ejection fraction (LVEF) was used to assess LV function. All patients underwent dynamic ECG and laboratory evaluations. This trial is registered with ClinicalTrails.gov, number NCT04421274. RESULTS Between March 2008 and July 2010, 43 patients who had underwent optimum medical treatment and successful percutaneous coronary intervention were randomly assigned to BM-MSC group (n = 21) or control group (n = 22) and followed-up for 12 months. At the 6th month's follow-up, there was no significant improvement in myocardial activity in the BM-MSC group before and after transplantation. Meanwhile, there was no statistically significant difference between the two groups in the change of myocardial perfusion defect index (p = 0.37) and myocardial metabolic defect index (p = 0.90). The LVEF increased from baseline to 12 months in the BM-MSC group and control group (mean baseline-adjusted BM-MSC treatment differences in LVEF 4.8% (SD 9.0) and mean baseline-adjusted control group treatment differences in LVEF 5.8% (SD 6.04)). However, there was no statistically significant difference between the two groups in the change of the LVEF (p = 0.23). We noticed that during the 12 months' follow-up, except for one death and one coronary microvascular embolism in the BM-MSC group, no other events occurred and alanine transaminase (ALT) and C-reactive protein (CRP) in BM-MSC group were significantly lower than that in the control group. CONCLUSIONS The present study may have many methodological limitations, and within those limitations, we did not identify that intracoronary transfer of autologous BM-MSCs could largely promote the recovery of LV function and myocardial viability after acute myocardial infarction.
Collapse
Affiliation(s)
- Runfeng Zhang
- Department of Cardiology, Department of Clinical Pharmacy, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Jiang Yu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ningkun Zhang
- Heart Centre, The Navy General Hospital, Beijing, 100048, China
| | - Wensong Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jisheng Wang
- Department of Cardiology, Department of Clinical Pharmacy, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Guocai Cai
- Department of Cardiology, Department of Clinical Pharmacy, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Yu Chen
- Heart Centre, The Navy General Hospital, Beijing, 100048, China
| | - Yong Yang
- Department of Cardiology, The General Hospital of Chinese People's Armed Police Forces, Beijing, 100039, China
| | - Zhenhong Liu
- Department of Cardiology, Department of Clinical Pharmacy, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China.
| |
Collapse
|
37
|
Campos de Carvalho AC, Kasai-Brunswick TH, Bastos Carvalho A. Cell-Based Therapies for Heart Failure. Front Pharmacol 2021; 12:641116. [PMID: 33912054 PMCID: PMC8072383 DOI: 10.3389/fphar.2021.641116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/11/2021] [Indexed: 02/05/2023] Open
Abstract
Heart failure has reached epidemic proportions with the advances in cardiovascular therapies for ischemic heart diseases and the progressive aging of the world population. Efficient pharmacological therapies are available for treating heart failure, but unfortunately, even with optimized therapy, prognosis is often poor. Their last therapeutic option is, therefore, a heart transplantation with limited organ supply and complications related to immunosuppression. In this setting, cell therapies have emerged as an alternative. Many clinical trials have now been performed using different cell types and injection routes. In this perspective, we will analyze the results of such trials and discuss future perspectives for cell therapies as an efficacious treatment of heart failure.
Collapse
Affiliation(s)
- Antonio Carlos Campos de Carvalho
- Laboratory of Cellular and Molecular Cardiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Antonio Carlos Campos de Carvalho,
| | - Tais H. Kasai-Brunswick
- National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriana Bastos Carvalho
- Laboratory of Cellular and Molecular Cardiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology in Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
38
|
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]
|
39
|
Affiliation(s)
- Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
40
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
41
|
Fan X, Li K, Zhu L, Deng X, Feng Z, Xu C, Liu S, Wu J. Prolonged therapeutic effects of photoactivated adipose-derived stem cells following ischaemic injury. Acta Physiol (Oxf) 2020; 230:e13475. [PMID: 32306486 DOI: 10.1111/apha.13475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/31/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
Abstract
AIM Adipose-derived stem cells (ASCs) therapies are emerging as a promising approach to therapeutic angiogenesis. Therapeutic persistence and reduced primitive stem cell function following cell delivery remains a critical hurdle for the clinical translation of stem cells in current approaches. METHODS Cultured ASCs were derived from subcutaneous white adipose tissue isolated from mice fed a normal diet (ND). Unilateral hindlimb ischaemia model was induced in high-fat diet (HFD)-fed mice by femoral artery interruption, after which photoactivated and non-light-treated ASCs were injected into the tail vein of mice. Laser Doppler imaging was conducted to measure the blood flow reperfusion. Capillary density was measured in the ischaemic gastrocnemius muscle. mRNA levels of angiogenic factors were determined by reverse-transcription polymerase chain reaction. Flow cytometry was used to determine the characterization of ASCs and endothelial progenitor cell (EPC). Human ASCs secretomes were analysed by liquid chromatography tandem mass spectrometry. RESULTS Our study demonstrated that photoactivated ND-ASCs prolonged functional blood flow perfusion and increased ASCs-derived EPC and neovascularization 38 days after ligation, when compared with saline-treated controls. Profiling analysis in ischaemic muscles showed upregulation of genes associated with pro-angiogenic factors after injection of photoactivated ND-ASCs when compared with the non-light-treated ASCs or saline treated HFD mice. Mass spectrometry revealed that light-treated ASCs conditioned medium retained a more complete pro-angiogenic activity with significant upregulation of angiogenesis related proteins. CONCLUSION Our data demonstrates that photoactivated ND-ASCs improve blood flow recovery and their injection may prove to be a useful strategy for the prevention and treatment of diabetic peripheral arterial disease.
Collapse
Affiliation(s)
- Xin Fan
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Kai Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Luochen Zhu
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Xin Deng
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Ziqian Feng
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Chunrong Xu
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Sijing Liu
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province Drug Discovery Research Center Southwest Medical University Luzhou China
- Laboratory for Cardiovascular Pharmacology Department of Pharmacology School of Pharmacy Southwest Medical University Luzhou China
| |
Collapse
|
42
|
Gastl M, Sürder D, Corti R, Faruque Osmany D, Gotschy A, von Spizcak J, Sokolska J, Metzen D, Alkadhi H, Ruschitzka F, Kozerke S, Manka R. Effect of intracoronary bone marrow-derived mononuclear cell injection early and late after myocardial infarction on CMR-derived myocardial strain. Int J Cardiol 2020; 310:108-115. [DOI: 10.1016/j.ijcard.2020.01.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/20/2019] [Accepted: 01/13/2020] [Indexed: 11/24/2022]
|
43
|
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: 16] [Impact Index Per Article: 3.2] [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
|
44
|
Fine B, Vunjak-Novakovic G. Heart regeneration in mouse and human: A bioengineering perspective. CURRENT OPINION IN PHYSIOLOGY 2020; 14:56-63. [PMID: 32095673 DOI: 10.1016/j.cophys.2020.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this short review, we draw parallels and stress differences between heart regeneration in mice and human, from a bioengineering perspective. As the prevailing dogma that the adult heart is completely post-mitotic is starting to change, there are multiple opportunities for augmenting the limited but definitive turnover of cardiomyocytes, to the extent necessary developing clinically relevant modalities for enhancing heart repair. We discuss some of the most promising among these new directions: mobilization of paracrine signaling by therapeutic cells, cell-free therapy of the heart using extracellular vesicles, and direct reprograming of endogenous cells. These new directions share the cell-free, mechanistic approach to heart repair that could be translated into the clinic faster and safer than the traditional cell therapies.
Collapse
Affiliation(s)
- Barry Fine
- Department of Medicine, Engineering Columbia University, New York NY 10032
| | - Gordana Vunjak-Novakovic
- Department of Medicine, Engineering Columbia University, New York NY 10032.,Department of Biomedical Engineering Columbia University, New York NY 10032
| |
Collapse
|
45
|
Peregud-Pogorzelska M, Przybycień K, Baumert B, Kotowski M, Pius-Sadowska E, Safranow K, Peregud-Pogorzelski J, Kornacewicz-Jach Z, Paczkowska E, Machaliński B. The Effect of Intracoronary Infusion of Autologous Bone Marrow-Derived Lineage-Negative Stem/Progenitor Cells on Remodeling of Post-Infarcted Heart in Patient with Acute Myocardial Infarction. Int J Med Sci 2020; 17:985-994. [PMID: 32410827 PMCID: PMC7211150 DOI: 10.7150/ijms.42561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: Regenerative capacity of the heart is limited, and the post-infarct left ventricle (LV) dysfunction is associated with poor prognosis. Administration of stem/progenitor cells (SPCs) is a promising approach for cardiac regeneration. Objectives: In the study, we assessed LV function and post-infarcted remodeling in patients with ST-elevated myocardial infarct (STEMI) who received autologous lineage-negative (LIN-) SPCs. Patients and methods: Patients with STEMI and one-vessel coronary artery disease treated with percutaneous revascularisation were divided into study group (LIN- group, 15 patients) that received standard therapy and autologous BM-derived LIN- SPCs and control group (standard therapy group, 19 patients). The cells were administered intracoronary 24 hours after STEMI. The follow-up was 12 months with subsequent non-invasive tests and laboratory parameter evaluation on days 1st, 3rd, and 7th as well as at 1st, 3rd, 6th and 12th month after STEMI. Results: All procedures related to SPCs administration were well tolerated by the patients. In 12-month follow-up, there were no major adverse cardiac events connected with LIN- SPCs administration. During 12-month follow-up, 9 patients from LIN- group (Responders) achieved an improvement in LV ejection fraction (>10% after 12 months) with no signs of unfavorable LV remodeling. Laboratory parameters analysis showed that Troponin T levels were significantly lower until day 7th in the Responders group, while brain natriuretic peptide (BNP) level remained significantly lower from day 3rd to 12th month respectively. Conclusions: Intracoronary infusion of autologous BM-derived LIN- stem/progenitor cells is feasible and safe for patient. Improvement in LV function and prevention of unfavorable remodeling in the 60% of study group seems relatively promising. Stem cell-based therapy for cardiac regeneration still needs more accurate and extensive investigations to estimate and improve their efficacy.
Collapse
Affiliation(s)
| | - Krzysztof Przybycień
- Department of Cardiology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Bartłomiej Baumert
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Maciej Kotowski
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | | | | | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
46
|
Ruhatiya RS, Adukia SA, Manjunath RB, Maheshwarappa HM. Current Status and Recommendations in Multimodal Neuromonitoring. Indian J Crit Care Med 2020; 24:353-360. [PMID: 32728329 PMCID: PMC7358870 DOI: 10.5005/jp-journals-10071-23431] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Every patient in neurocritical care evolves through two phases. Acute pathologies are addressed first. These include trauma, hemorrhagic or ischemic stroke, or neuroinfection. Soon after, the concentration shifts to identifying secondary pathologies like fever, seizures, and ischemia, which may exacerbate the brain injury. Frequent bedside examinations are not sufficient for timely detection and prevention of secondary brain injury (SBI) as per the International Multidisciplinary Consensus Conference on Multimodality Monitoring in Neurocritical Care. Multimodality monitoring (MMM) can help in tailoring treatment decisions to prevent such a brain injury. Multimodal neuromonitoring involves data-guided therapeutic interventions by employing various tools and data integration to understand brain physiology. Monitors provide real-time information on cerebral hemodynamics, oxygenation, metabolism, and electrophysiology. The monitors may be invasive/noninvasive and global/regional. We have reviewed such technologies in this write-up. Novel themes like bioinformatics, clinical research, and device development will also be discussed.
Collapse
Affiliation(s)
- Radhika S Ruhatiya
- Department of Critical Care Medicine, Narayana Hrudayalaya, NH Health City, Bengaluru, Karnataka, India
| | - Sachin A Adukia
- Department of Neurology, Narayana Hrudayalaya, NH Health City, Bengaluru, Karnataka, India
| | - Ramya B Manjunath
- Department of Anesthesia, Narayana Hrudayalaya, NH Health City, Bengaluru, Karnataka, India
| | - Harish M Maheshwarappa
- Department of Critical Care Medicine, Narayana Hrudayalaya, NH Health City, Bengaluru, Karnataka, India
| |
Collapse
|
47
|
Xu JY, Qian HY, Huang PS, Xu J, Xiong YY, Jiang WY, Xu Y, Leng WX, Li XD, Chen GH, Tang RJ, Huang CR, Hu MJ, Jin C, Wu Y, Zhang J, Qian J, Xu B, Zhao SH, Lu MJ, Shen R, Fang W, Wu WC, Chen X, Wang Y, Li W, Lu XF, Jiang XF, Ma CC, Li JW, Geng YJ, Qiao SB, Gao RL, Yang YJ. Transplantation efficacy of autologous bone marrow mesenchymal stem cells combined with atorvastatin for acute myocardial infarction (TEAM-AMI): rationale and design of a randomized, double-blind, placebo-controlled, multi-center, Phase II TEAM-AMI trial. Regen Med 2019; 14:1077-1087. [PMID: 31829095 DOI: 10.2217/rme-2019-0024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: To determine the efficacy and safety of intracoronary infusion of autologous bone marrow mesenchymal stem cells (MSCINJ) in combination with intensive atorvastatin (ATV) treatment for patients with anterior ST-segment elevation myocardial infarction-elevation myocardial infarction. Patients & methods: The trial enrolls a total of 100 patients with anterior ST-elevation myocardial infarction. The subjects are randomly assigned (1:1:1:1) to receive routine ATV (20 mg/d) with placebo or MSCsINJ and intensive ATV (80 mg/d) with placebo or MSCsINJ. The primary end point is the absolute change of left ventricular ejection fraction within 12 months. The secondary end points include parameters in cardiac function, remodeling and regeneration, quality of life, biomarkers and clinical outcomes. Results & conclusion: The trial will implicate the essential of cardiac micro-environment improvement (‘fertilizing’) for cell-based therapy. Clinical Trial Registration: NCT03047772.
Collapse
Affiliation(s)
- Jun-Yan Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Hai-Yan Qian
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Pei-Sen Huang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Jun Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Yu-Yan Xiong
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Wen-Yang Jiang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Yi Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Wen-Xiu Leng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Xiang-Dong Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Gui-Hao Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Rui-Jie Tang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Cun-Rong Huang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Meng-Jin Hu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Chen Jin
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Yuan Wu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Jun Zhang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Jie Qian
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Bo Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Shi-Hua Zhao
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Min-Jie Lu
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Rui Shen
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Wei Fang
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Wei-Chun Wu
- Department of Echocardiography, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Xi Chen
- Center of Laboratory Medicine, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Yang Wang
- Medical Research & Biometrics Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Wei Li
- Medical Research & Biometrics Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Xiang-Feng Lu
- Department of Epidemiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Xi-Feng Jiang
- Hebei Better Cell Biological Technology Co., Ltd, Hebei 050000, China
| | - Chun-Cheng Ma
- Hebei Better Cell Biological Technology Co., Ltd, Hebei 050000, China
| | - Jian-Wen Li
- Hebei Better Cell Biological Technology Co., Ltd, Hebei 050000, China
| | - Yong-Jian Geng
- The Center for Cardiovascular Biology & Atherosclerosis Research, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Shu-Bin Qiao
- Hebei Better Cell Biological Technology Co., Ltd, Hebei 050000, China
| | - Run-Lin Gao
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | - Yue-Jin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
48
|
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
|
49
|
Xie DM, Li YL, Li J, Li Q, Lu G, Zhai Y, Zhang J, Huang Z, Gao X. CD51 distinguishes a subpopulation of bone marrow mesenchymal stem cells with distinct migratory potential: a novel cell-based strategy to treat acute myocardial infarction in mice. Stem Cell Res Ther 2019; 10:331. [PMID: 31747966 PMCID: PMC6865070 DOI: 10.1186/s13287-019-1439-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 11/22/2022] Open
Abstract
Background Experimental and clinical trials have demonstrated the efficiency of bone marrow-derived mesenchymal stromal/stem cells (bMSCs) in the treatment of myocardial infarction. However, after intravenous injection, the ineffective migration of engrafted bMSCs to the hearts remains an obstacle, which has an undesirable impact on the efficiency of cell-based therapy. Therefore, we attempted to identify a marker that could distinguish a subpopulation of bMSCs with a promising migratory capacity. Methods Here, CD51-negative and CD51-positive cells were isolated by flow cytometry from Ter119−CD45−CD31−bMSCs and cultured in specifically modified medium. The proliferation ability of the cells was evaluated by 5-ethynyl-2′-deoxyuridine (EdU) staining or continuously monitored during culture, and the differentiation potential was assessed by culturing the cells in the appropriate conditioned media. Wound healing assays, transwell assays and quantitative polymerase chain reaction (qPCR) were used to measure the migratory ability. The mice were subjected to a sham operation or myocardial infarction (MI) by permanently occluding the coronary artery, and green fluorescent protein (GFP)-labelled cells were transplanted into the mice via intravenous infusion immediately after MI. Heart function was measured by echocardiography; infarct myocardium tissues were detected by triphenyl tetrazolium chloride (TTC) staining. Additionally, immunofluorescence staining was used to verify the characteristics of CD51+bMSCs and inflammatory responses in vivo. Statistical comparisons were performed using a two-tailed Student’s t test. Results In this study, the isolated CD51−bMSCs and CD51+bMSCs, especially the CD51+ cells, presented a favourable proliferative capacity and could differentiate into adipocytes, osteocytes and chondrocytes in vitro. After the cells were transplanted into the MI mice by intravenous injection, the therapeutic efficiency of CD51+bMSCs in improving left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) was better than that of CD51−bMSCs. Compared with CD51−bMSCs, CD51+bMSCs preferentially migrated to and were retained in the infarcted hearts at 48 h and 8 days after intravenous injection. Accordingly, the migratory capacity of CD51+bMSCs exceeded that of CD51−bMSCs in vitro, and the former cells expressed higher levels of chemokine receptors or ligands. Interestingly, the retained CD51+bMSCs retained in the myocardium possessed proliferative potential but only differentiated into endothelial cells, smooth muscle cells, fibroblasts or cardiomyocytes. Transplantation of CD51+bMSCs partially attenuated the inflammatory response in the hearts after MI, while the potential for inflammatory suppression was low in CD51−bMSC-treated mice. Conclusions These findings indicated that the CD51-distinguished subpopulation of bMSCs facilitated proliferation and migration both in vitro and in vivo, which provided a novel cell-based strategy to treat acute MI in mice by intravenous injection.
Collapse
Affiliation(s)
- Dong-Mei Xie
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China
| | - Yuan-Long Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jie Li
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China
| | - Qinglang Li
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China
| | - Guihua Lu
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China
| | - Yuansheng Zhai
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China
| | - Juhong Zhang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China
| | - Zhibin Huang
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Xiuren Gao
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China. .,NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, 510080, China.
| |
Collapse
|
50
|
Salazar-Noratto GE, Luo G, Denoeud C, Padrona M, Moya A, Bensidhoum M, Bizios R, Potier E, Logeart-Avramoglou D, Petite H. Understanding and leveraging cell metabolism to enhance mesenchymal stem cell transplantation survival in tissue engineering and regenerative medicine applications. Stem Cells 2019; 38:22-33. [PMID: 31408238 DOI: 10.1002/stem.3079] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/06/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022]
Abstract
In tissue engineering and regenerative medicine, stem cell-specifically, mesenchymal stromal/stem cells (MSCs)-therapies have fallen short of their initial promise and hype. The observed marginal, to no benefit, success in several applications has been attributed primarily to poor cell survival and engraftment at transplantation sites. MSCs have a metabolism that is flexible enough to enable them to fulfill their various cellular functions and remarkably sensitive to different cellular and environmental cues. At the transplantation sites, MSCs experience hostile environments devoid or, at the very least, severely depleted of oxygen and nutrients. The impact of this particular setting on MSC metabolism ultimately affects their survival and function. In order to develop the next generation of cell-delivery materials and methods, scientists must have a better understanding of the metabolic switches MSCs experience upon transplantation. By designing treatment strategies with cell metabolism in mind, scientists may improve survival and the overall therapeutic potential of MSCs. Here, we provide a comprehensive review of plausible metabolic switches in response to implantation and of the various strategies currently used to leverage MSC metabolism to improve stem cell-based therapeutics.
Collapse
Affiliation(s)
- Giuliana E Salazar-Noratto
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Guotian Luo
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Cyprien Denoeud
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Mathilde Padrona
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Adrien Moya
- South Florida Veterans Affairs Foundation for Research and Education, Inc., Miami, Florida.,Geriatric Research, Education and Clinical Center and Research Service, Bruce W. Carter VAMC, Miami, Florida
| | - Morad Bensidhoum
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Rena Bizios
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, Texas
| | - Esther Potier
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Delphine Logeart-Avramoglou
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| | - Hervé Petite
- Université de Paris, B3OA CNRS INSERM, Paris, France.,Ecole Nationale Vétérinaire d'Alfort, B3OA, Maisons-Alfort, France
| |
Collapse
|