1
|
Pan M, Xu Y, Wang Y, Jiang Y, Xie Y, Tai C, Wang W, Wang B. The therapeutic efficacy comparison of MSCs derived different tissues unveilings anti-apoptosis more crucial than angiogenesis in treating acute myocardial infarction. Stem Cell Res Ther 2025; 16:236. [PMID: 40361236 PMCID: PMC12077008 DOI: 10.1186/s13287-025-04378-3] [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] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Myocardial infarction (MI) is a severe disease that often associated with impaired angiogenesis and increased myocardial apoptosis. Mesenchymal stromal cells (MSCs) have been a promising candidate for treating myocardial infarction. However, functional heterogeneity of MSCs leads to inconsistent therapeutic efficiency and the current MSCs-based therapy lacks the concept and implementation of precision medicine. In this study, we compared the cardioprotective effect of UCMSCs and ADMSCs targeting the angiogenesis in a mouse MI model and screened out optimum MSCs candidate for precise clinical application. METHODS The gene expression profiles of UCMSCs and ADMSCs were investigated through RNA sequencing analysis. To compare their angiogenic potential, we performed tube formation assay, Matrigel plug assays, and aortic ring assay, and analyzed pro-angiogenic genes via qPCR. Subsequently, UCMSCs and ADMSCs were respectively injected into myocardium after MI surgery in mice. On day 28 post-MI, echocardiography was performed to assess cardiac function. Histological analysis was performed to assess MSCs retention, angiogenesis, and myocardial apoptosis. Additionally, the anti-apoptosis effects mediated by MSCs were further evaluated using flow cytometry in hypoxia H9C2 and HL-1 cells. RESULTS The RNA sequencing analysis revealed differences in gene expression related to angiogenesis and apoptosis pathways between UCMSCs and ADMSCs. UCMSCs presented greater pro-angiogenesis activity than ADMSCs in vitro and in vivo. Both of UCMSCs and ADMSCs improved cardiac function, decreased infarction area and inhibited cardiomyocyte apoptosis while promoting angiogenesis post-MI in mice. Notably, ADMSCs exerted a better cardioprotective function than UCMSCs and stronger anti-apoptotic effect on residual cardiomyocytes. CONCLUSIONS The protection of residual cells survival played a more prominent role than angiogenesis in MSCs-based therapy for acute MI. Our study provides new insights into therapeutic strategies and suggests that the optimal type of MSCs can be screened based on their tissue heterogeneity for precise clinical applications in acute MI.
Collapse
Affiliation(s)
- Mingjie Pan
- Clinical Medicine Research Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yueyue Xu
- The Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yaping Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital, Clinical Medical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yue Jiang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yuanyuan Xie
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Chenxu Tai
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Wenqing Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, Nanjing Drum Tower Hospital, Clinical Medical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
| |
Collapse
|
2
|
ZHANG SY. Chinese Guidelines for the Diagnosis and Treatment of Heart Failure 2024. J Geriatr Cardiol 2025; 22:277-331. [PMID: 40351394 PMCID: PMC12059564 DOI: 10.26599/1671-5411.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
In the past 6 years, significant breakthroughs have been achieved in the treatment of heart failure (HF), especially in drug therapy. The classification of chronic HF and the treatment methods for HF and its complications are also constantly being updated. In order to apply these results to the diagnosis and treatment of patients with HF in China and further improve the level of diagnosis and treatment of HF in China, the HF Group of Chinese Society of Cardiology, Chinese Medical Association, Chinese College of Cardiovascular Physician, Chinese HF Association of Chinese Medical Doctor Association, and Editorial Board of Chinese Journal of Cardiology have organized an expert group and update the consensus and evidence-based treatment methods in the field of HF based on the latest clinical research findings at home and abroad, combined with the national conditions and clinical practice in China, and referring to the latest foreign HF guidelines while maintaining the basic framework of the 2018 Chinese Guidelines for Diagnosis and Treatment of HF.
Collapse
Affiliation(s)
- Shu-Yang ZHANG
- Chinese Society of Cardiology, Chinese Medical Association; Chinese College of Cardiovascular Physician; Chinese Heart Failure Association of Chinese Medical Doctor Association; Editorial Board of Chinese Journal of Cardiology;This guideline was first published in the Zhonghua Xin Xue Guan Bing Za Zhi 2024; 52(3): 235–275.
| |
Collapse
|
3
|
Almeida M, Inácio JM, Vital CM, Rodrigues MR, Araújo BC, Belo JA. Cell Reprogramming, Transdifferentiation, and Dedifferentiation Approaches for Heart Repair. Int J Mol Sci 2025; 26:3063. [PMID: 40243729 PMCID: PMC11988544 DOI: 10.3390/ijms26073063] [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: 03/01/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death globally, with myocardial infarction (MI) being a major contributor. The current therapeutic approaches are limited in effectively regenerating damaged cardiac tissue. Up-to-date strategies for heart regeneration/reconstitution aim at cardiac remodeling through repairing the damaged tissue with an external cell source or by stimulating the existing cells to proliferate and repopulate the compromised area. Cell reprogramming is addressed to this challenge as a promising solution, converting fibroblasts and other cell types into functional cardiomyocytes, either by reverting cells to a pluripotent state or by directly switching cell lineage. Several strategies such as gene editing and the application of miRNA and small molecules have been explored for their potential to enhance cardiac regeneration. Those strategies take advantage of cell plasticity by introducing reprogramming factors that regress cell maturity in vitro, allowing for their later differentiation and thus endorsing cell transplantation, or promote in situ cell proliferation, leveraged by scaffolds embedded with pro-regenerative factors promoting efficient heart restoration. Despite notable advancements, important challenges persist, including low reprogramming efficiency, cell maturation limitations, and safety concerns in clinical applications. Nonetheless, integrating these innovative approaches offers a promising alternative for restoring cardiac function and reducing the dependency on full heart transplants.
Collapse
Affiliation(s)
| | - José M. Inácio
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (M.A.); (C.M.V.); (M.R.R.); (B.C.A.)
| | | | | | | | - José A. Belo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal; (M.A.); (C.M.V.); (M.R.R.); (B.C.A.)
| |
Collapse
|
4
|
Zhong X, Luo L, Wu J, Li W, Liu X, Ye T, Li Z, Shi P. Adhesion-Assisted Antioxidant-Engineered Mesenchymal Stromal Cells for Enhanced Cardiac Repair in Myocardial Infarction. ACS NANO 2025; 19:11412-11426. [PMID: 40073336 DOI: 10.1021/acsnano.5c00820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Mesenchymal stromal cell (MSC) therapy holds great promise for treating myocardial infarction (MI). However, the inflammatory and reactive oxygen species (ROS)-rich environment in infarcted myocardium challenges MSC survival, limiting its therapeutic impact. In this study, we demonstrate that chemical modification of MSCs with anti-VCAM1 and polydopamine (PD) significantly enhances MSC survival and promotes cardiac repair. Anti-VCAM1 modification facilitates MSC adhesion to inflamed tissue, ensuring MSC retention in the injured myocardium, while PD scavenges ROS surrounding MSCs, creating a conducive environment for cell transplantation. Our data indicate that chemically engineered MSCs effectively disrupt the inflammation-ROS cycle and modulate inflammation-related immune responses, thus improving MI microenvironments. Single-cell RNA sequencing of rat hearts reveals that treatment with engineered MSCs inhibits cardiac fibrosis by suppressing HB-EGF-EGFR signaling between anti-inflammatory macrophages and activated fibrillates. Ultimately, engineered MSCs demonstrate superior therapeutic efficacy in a rat model of MI. This study presents a straightforward, safe, and efficient chemical method for enhancing MSC therapy.
Collapse
Affiliation(s)
- Xianghua Zhong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Li Luo
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, P. R. China
| | - Jiyuan Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Weirun Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, P. R. China
| | - Xinyang Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Tenghui Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Zhenhua Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, P. R. China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
5
|
Umrath F, Frick SL, Wendt V, Naros A, Zimmerer R, Alexander D. Inhibition of TGF-β signaling enhances osteogenic potential of iPSC-derived MSCs. Sci Rep 2025; 15:7814. [PMID: 40050624 PMCID: PMC11885616 DOI: 10.1038/s41598-025-89370-w] [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] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Mesenchymal stem cells (MSCs) represent the most commonly utilized type of stem cell in clinical applications. However, variability in quality and quantity between different tissue sources and donors presents a significant challenge to their use. Induced pluripotent stem cells (iPSCs) are a promising and abundant alternative source of MSCs, offering a potential solution to the limitations of adult MSCs. Nevertheless, a standardized protocol for the differentiation of iPSCs into iPSC-derived mesenchymal stem cells (iMSCs) has yet to be established, as the existing methods vary significantly in terms of complexity, duration, and outcome. Many straightforward methods induce differentiation by culturing iPSCs in MSC media which are supplemented with fetal bovine serum (FBS) or human platelet lysate (hPL), followed by selection of MSC-like cells by passaging. However, in our hands, this approach yielded inconsistent quality of iMSCs, particularly in terms of osteogenic potential and premature senescence. This study examines the impact of the selective TGF-β inhibitor SB431542 on iMSC differentiation, demonstrating that TGF-β inhibition enhances osteogenic potential and reduces premature senescence. Additionally, we present a reliable, xeno-free method for producing high-quality iMSCs that can be adapted for Good Manufacturing Practice (GMP) compliance, thus enhancing the potential for clinical applications.
Collapse
Affiliation(s)
- Felix Umrath
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstr. 2-8, Tübingen, 72076, Germany.
- Department of Orthopedic Surgery, University Hospital Tübingen, Tübingen, Germany.
| | - Sarah-Lena Frick
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstr. 2-8, Tübingen, 72076, Germany
| | - Valerie Wendt
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstr. 2-8, Tübingen, 72076, Germany
| | - Andreas Naros
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstr. 2-8, Tübingen, 72076, Germany
| | - Rüdiger Zimmerer
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstr. 2-8, Tübingen, 72076, Germany
| | - Dorothea Alexander
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Osianderstr. 2-8, Tübingen, 72076, Germany
| |
Collapse
|
6
|
Liu H, Xu Y, Liu Y, Han X, Zhao L, Liu Y, Zhang F, Fu Y. Identification of novel ferroptosis-related biomarkers associated with the oxidative stress pathways in ischemic cardiomyopathy. IJC HEART & VASCULATURE 2025; 56:101584. [PMID: 39807364 PMCID: PMC11726793 DOI: 10.1016/j.ijcha.2024.101584] [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: 11/04/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Background Ferroptosis is a cell death process that depends on iron and reactive oxygen species. It significantly contributes to cardiovascular diseases. However, its exact role in ischemic cardiomyopathy (ICM) is still unclear. Methods Using bioinformatics methods, we identified new molecular targets associated with ferroptosis in ICM and conducted various analyses-including correlation analysis, pathway enrichment analysis, protein interaction network construction, and analysis of transcription factor and drug interactions, to reveal the potential mechanisms behind these genes. Results We evaluated two independent training sets of ICM, GSE57338 and GSE5406, comprising 203 ICM samples, and validation sets GSE76701 to examine differentially expressed genes (DEGs) related to ferroptosis. After extracting the intersection of the gene sets and ferroptosis-related genes, 53 DEGs were identified. Enrichment analyses showed that the alterations in ferroptosis-related DEGs were mainly enriched in oxidative stress response, and immune-related pathways. Furthermore, 11 hub genes were identified using protein-protein interaction network analysis. The key interactions between 11 hub genes were more pronounced in protein localization during ICM development. In addition, we construct a hub gene and transcription factor interaction network and a small molecule drug-gene interaction network. We found that among these hub genes, the N-acetylneuraminate outer membrane channel(NANC) gene is positively correlated with most of the small-molecule drugs used to treat ICM, and its high expression might increase resistance. Conclusions Ferroptosis exists in ICM and and is associated with oxidative stress. This association suggests that ferroptosis may facilitate the progression of ICM.
Collapse
Affiliation(s)
- Huilin Liu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - Yuan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yuanmei Liu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - XueJun Han
- Department of Orthopaedics, Jiayuguan Municipal First People’s Hospital, Jiayuguan 735100, PR China
| | - Liping Zhao
- Department of Ophthalmology, Jiayuguan Municipal First People’s Hospital, Jiayuguan 735100, PR China
| | - Yixuan Liu
- College of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Fuchun Zhang
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| | - Yicheng Fu
- Department of Geriatrics, Peking University Third Hospital, Beijing 100191, PR China
| |
Collapse
|
7
|
He X, Good A, Kalou W, Ahmad W, Dutta S, Chen S, Lin CN, Chella Krishnan K, Fan Y, Huang W, Liang J, Wang Y. Current Advances and Future Directions of Pluripotent Stem Cells-Derived Engineered Heart Tissue for Treatment of Cardiovascular Diseases. Cells 2024; 13:2098. [PMID: 39768189 PMCID: PMC11674482 DOI: 10.3390/cells13242098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular diseases resulting from myocardial infarction (MI) remain a leading cause of death worldwide, imposing a substantial burden on global health systems. Current MI treatments, primarily pharmacological and surgical, do not regenerate lost myocardium, leaving patients at high risk for heart failure. Engineered heart tissue (EHT) offers a promising solution for MI and related cardiac conditions by replenishing myocardial loss. However, challenges like immune rejection, inadequate vascularization, limited mechanical strength, and incomplete tissue maturation hinder clinical application. The discovery of human-induced pluripotent stem cells (hiPSCs) has transformed the EHT field, enabling new bioengineering innovations. This review explores recent advancements and future directions in hiPSC-derived EHTs, focusing on innovative materials and fabrication methods like bioprinting and decellularization, and assessing their therapeutic potential through preclinical and clinical studies. Achieving functional integration of EHTs in the heart remains challenging due to the need for synchronized contraction, sufficient vascularization, and mechanical compatibility. Solutions such as genome editing, personalized medicine, and AI technologies offer promising strategies to address these translational barriers. Beyond MI, EHTs also show potential in treating ischemic cardiomyopathy, heart valve engineering, and drug screening, underscoring their promise in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Angela Good
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Wael Kalou
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Waqas Ahmad
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Sophie Chen
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Charles Noah Lin
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yanbo Fan
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Wei Huang
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| |
Collapse
|
8
|
Cartuliares MB, Hejbøl EK, Schrøder HD, Pedersen AK, Frich LH. Stem cell treatment for regeneration of the rotator cuff: study protocol for a prospective single-center randomized controlled trial (Lipo-cuff). Trials 2024; 25:696. [PMID: 39427182 PMCID: PMC11492208 DOI: 10.1186/s13063-024-08557-0] [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: 09/02/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Rotator cuff tears (RCT) are a common musculoskeletal condition, especially in the aging population. The prevalence of rotator cuff tears varies based on factors like age, occupation, and activity level. In the general population, the prevalence of rotator cuff tears is estimated to be around 20 to 25%. Rotator cuff tears (RCT) have an impact in patients' pain level, shoulder function, sleep disturbance, and quality of life. Primary tendon surgery is in mostly cases necessary. This study aimed to examine if treatment of rotator cuff lesions with implantation of micro-fragmented adipose tissue can improve patients' reported pain and function compared to conventional surgery. METHODS The study is a prospective superiority parallel-group single-center randomized controlled trial including 30 patients between 40 and 69 years of age in Denmark. Patients will be allocated 1:1 ratio to reconstruction of the supraspinatus tendon with an injection of micro-fragmented adipose tissue into the related muscle (stem cell treatment) or the standard of care (SOC), which is conventional surgery. Patients, project assistants, physicians, and outcome adjudicators are not blinded to randomization due to practical constraints. The radiologist and the statistician performing the analysis will be blinded. The primary outcome will be the Oxford shoulder score at 12 months post-surgery. DISCUSSION This study will assess whether adding micro-fragmented adipose tissue therapy to conventional rotator cuff tear treatment can enhance recovery, accelerate return to daily activities, and improve functional outcomes. The research will also determine if this minimally invasive procedure could be standardized for routine patient care. TRIAL REGISTRATION ClinicalTrials.gov NCT06505135. Registered on July 10, 2024.
Collapse
Affiliation(s)
- Mariana Bichuette Cartuliares
- Department of Orthopaedics, Hospital Soenderjylland, Kresten Philipsens Vej 15, Aabenraa, 6200, Denmark.
- Department of Regional Health Research, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark.
| | - Eva Kildall Hejbøl
- Department of Regional Health Research, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark
- Department of Regional Health Research and Institute of Molecular Medicine, Orthopaedic Research Unit, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark
- Department of Pathology, Odense University Hospital, J. B. Winsløws Vej 15, Odense, 5000, Denmark
| | - Henrik Daa Schrøder
- Department of Pathology, Odense University Hospital, J. B. Winsløws Vej 15, Odense, 5000, Denmark
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, Odense, 5000, Denmark
| | - Andreas Kristian Pedersen
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, Odense, 5000, Denmark
- Research Unit OPEN - Open Patient Data Explorative Network, J. B. Winsløws Vej 21, Odense, 5000, Denmark
| | - Lars Henrik Frich
- Department of Orthopaedics, Hospital Soenderjylland, Kresten Philipsens Vej 15, Aabenraa, 6200, Denmark
- Department of Regional Health Research, University of Southern Denmark, Campusvej 55, Odense M, 5230, Denmark
| |
Collapse
|
9
|
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
|
10
|
Chacon Alberty L, King M, Mesquita FCP, Hochman-Mendez C. Quality Assessment of Long-Term Cryopreserved Human Bone-Derived Marrow Mesenchymal Stromal Cell Samples: Experience from the Texas Heart Institute Biorepository and Biospecimen Profiling Core. Biopreserv Biobank 2024. [PMID: 39253842 DOI: 10.1089/bio.2023.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
In biomedical research, biorepositories are pivotal resources that safeguard and supply clinical samples for scientific investigators. Proper long-term cryopreservation conditions are essential to maintain biospecimen quality. In this study, we analyzed the efficacy of sample cryopreservation at the Texas Heart Institute Biorepository and Biospecimen Profiling Core (THI-BRC). Our assessments included a thorough review of internal processes, quality reports, and both internal and external audit outcomes. We examined the integrity of human bone marrow-derived multipotent mesenchymal stromal cells (BM-MSCs) that were cryopreserved for over 5 years. These samples originated from randomly selected clinical trial participants or commercially sourced cell lines. Parameters such as cell viability, DNA and RNA integrity, population doubling time, sterility, and BM-MSC-specific attributes such as surface antigen expression and differentiation potential were studied. BM-MSC samples cryopreserved for ∼6 months served as our control. Our results demonstrated that the 5-year cryopreserved samples maintained their integrity compared with the shorter-term stored control samples. Moreover, THI-BRC has met accreditation agency standards and has not received any repeated deficiencies over 7 years. Collectively, our findings affirm that THI-BRC's biospecimen storage protocols align with accepted standards as confirmed by the quality assessment of long-term stored clinical samples.
Collapse
Affiliation(s)
- Lourdes Chacon Alberty
- Biorepository and Biospecimen Profiling Core, The Texas Heart Institute, Houston, Texas, USA
- Regenerative Medicine Research, The Texas Heart Institute, Houston, Texas, USA
| | - Madelyn King
- Biorepository and Biospecimen Profiling Core, The Texas Heart Institute, Houston, Texas, USA
| | | | - Camila Hochman-Mendez
- Biorepository and Biospecimen Profiling Core, The Texas Heart Institute, Houston, Texas, USA
- Regenerative Medicine Research, The Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
11
|
Lam J, Yu J, Lee B, Campagna C, Yoo S, Baek K, Jeon NL, Sung KE. Characterizing On-Chip Angiogenesis Induction in a Microphysiological System as a Functional Measure of Mesenchymal Stromal Cell Bioactivity. Adv Biol (Weinh) 2024; 8:e2300094. [PMID: 37409400 DOI: 10.1002/adbi.202300094] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/12/2023] [Indexed: 07/07/2023]
Abstract
Mesenchymal stromal cells (MSCs) continue to be proposed for clinical investigation to treat myriad diseases given their purported potential to stimulate endogenous regenerative processes, such as angiogenesis. However, MSC functional heterogeneity has hindered clinical success and still poses a substantial manufacturing challenge from a product quality control perspective. Here, a quantitative bioassay based on an enhanced-throughput is described, microphysiological system (MPS) to measure the specific bioactivity of MSCs to stimulate angiogenesis as a potential measure of MSC potency. Using this novel bioassay, MSCs derived from multiple donors at different passages are co-cultured with human umbilical vein endothelial cells and exhibit significant heterogeneity in angiogenic potency between donors and cell passage. Depending on donor source and cellular passage number, MSCs varied in their ability to stimulate tip cell dominant or stalk cell dominant phenotypes in angiogenic sprout morphology which correlated with expression levels of hepatocyte growth factor (HGF). These findings suggest that MSC angiogenic bioactivity may be considered as a possible potency attribute in MSC quality control strategies. Development of a reliable and functionally relevant potency assay for measuring clinically relevant potency attributes of MSCs will help to improve consistency in quality and thereby, accelerate clinical development of these cell-based products.
Collapse
Affiliation(s)
- Johnny Lam
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - James Yu
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Byungjun Lee
- Qureator, Inc., 7094 Miratech Drive, Suite 110, San Diego, CA, 92121, USA
| | - Courtney Campagna
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Sanghee Yoo
- Qureator, Inc., 7094 Miratech Drive, Suite 110, San Diego, CA, 92121, USA
| | - Kyusuk Baek
- Qureator, Inc., 7094 Miratech Drive, Suite 110, San Diego, CA, 92121, USA
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Kyung E Sung
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| |
Collapse
|
12
|
Masroor M, Wang Y, Zhang C, Dong N. Surgical Management of Ischemic Cardiomyopathy Patients with Severe Left Ventricular Dysfunction: Is It Time to Reconsider Revascularization Surgery? J Cardiovasc Dev Dis 2024; 11:184. [PMID: 39057607 PMCID: PMC11276879 DOI: 10.3390/jcdd11070184] [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: 04/20/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Ischemic cardiomyopathy patients with severe left ventricular dysfunction are a specific group of patients with poor surgical outcomes. There are few surgical treatment options in practice for the treatment of these patients such as heart transplantation, coronary artery bypass surgery, surgical ventricular restoration, etc. Despite multiple treatment options, there are no explicit clinical guidelines available to guide surgeons in choosing the most appropriate option and ensuring that the specific patient can benefit from the selected surgical treatment. Heart transplantation is the gold standard treatment for ischemic cardiomyopathy patients with severe left ventricular dysfunction, but it is limited to very few highly equipped centers around the world due to donor shortages, complex perioperative and surgical management, and limited technological and human resources. It is evident from some studies that heart transplant-eligible candidates can benefit from alternative surgical options such as coronary artery bypass surgery alone or combined with surgical ventricular restoration. Therefore, alternative surgical options that are used for most of the population, especially in developing and underdeveloped countries, need to be discussed to improve their outcomes. A challenge in the recent era which has yet to find a solution is to determine which heart transplant candidate can benefit from simple revascularization compared to a complex heart transplantation procedure. Myocardial viability testing was one of the most important determinants in deciding whether a patient should undergo revascularization, but its role in guiding appropriate surgical options has been challenged. This review aims to discuss the available surgical management options and their long-term outcomes for patients with ischemic cardiomyopathy, which will eventually help surgeons when choosing a surgical procedure.
Collapse
Affiliation(s)
- Matiullah Masroor
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
- Department of Cardiothoracic and Vascular Surgery, Amiri Medical Complex, Qargha Rd, Kabul 1010, Afghanistan
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
| | - Chao Zhang
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.M.); (Y.W.)
| |
Collapse
|
13
|
Akbar N, Razzaq SS, Salim A, Haneef K. Mesenchymal Stem Cell-Derived Exosomes and Their MicroRNAs in Heart Repair and Regeneration. J Cardiovasc Transl Res 2024; 17:505-522. [PMID: 37875715 DOI: 10.1007/s12265-023-10449-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023]
Abstract
Mesenchymal stem cells (MSCs) can be differentiated into cardiac, endothelial, and smooth muscle cells. Therefore, MSC-based therapeutic approaches have the potential to deal with the aftermaths of cardiac diseases. However, transplanted stem cells rarely survive in damaged myocardium, proposing that paracrine factors other than trans-differentiation may involve in heart regeneration. Apart from cytokines/growth factors, MSCs secret small, single-membrane organelles named exosomes. The MSC-secreted exosomes are enriched in lipids, proteins, nucleic acids, and microRNA (miRNA). There has been an increasing amount of data that confirmed that MSC-derived exosomes and their active molecule microRNA (miRNAs) regulate signaling pathways involved in heart repair/regeneration. In this review, we systematically present an overview of MSCs, their cardiac differentiation, and the role of MSC-derived exosomes and exosomal miRNAs in heart regeneration. In addition, biological functions regulated by MSC-derived exosomes and exosomal-derived miRNAs in the process of heart regeneration are reviewed.
Collapse
Affiliation(s)
- Nukhba Akbar
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Syeda Saima Razzaq
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
14
|
Zheng Z, Tang W, Li Y, Ai Y, Tu Z, Yang J, Fan C. Advancing cardiac regeneration through 3D bioprinting: methods, applications, and future directions. Heart Fail Rev 2024; 29:599-613. [PMID: 37943420 DOI: 10.1007/s10741-023-10367-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
Cardiovascular diseases (CVDs) represent a paramount global mortality concern, and their prevalence is on a relentless ascent. Despite the effectiveness of contemporary medical interventions in mitigating CVD-related fatality rates and complications, their efficacy remains curtailed by an array of limitations. These include the suboptimal efficiency of direct cell injection and an inherent disequilibrium between the demand and availability of heart transplantations. Consequently, the imperative to formulate innovative strategies for cardiac regeneration therapy becomes unmistakable. Within this context, 3D bioprinting technology emerges as a vanguard contender, occupying a pivotal niche in the realm of tissue engineering and regenerative medicine. This state-of-the-art methodology holds the potential to fabricate intricate heart tissues endowed with multifaceted structures and functionalities, thereby engendering substantial promise. By harnessing the prowess of 3D bioprinting, it becomes plausible to synthesize functional cardiac architectures seamlessly enmeshed with the host tissue, affording a viable avenue for the restitution of infarcted domains and, by extension, mitigating the onerous yoke of CVDs. In this review, we encapsulate the myriad applications of 3D bioprinting technology in the domain of heart tissue regeneration. Furthermore, we usher in the latest advancements in printing methodologies and bioinks, culminating in an exploration of the extant challenges and the vista of possibilities inherent to a diverse array of approaches.
Collapse
Affiliation(s)
- Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Weijie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yichen Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yinze Ai
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Zhi Tu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China.
| |
Collapse
|
15
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
16
|
Yuan HL, Chang L, Fan WW, Liu X, Li Q, Tian C, Zhao J, Li ZA, Pan XH, Zhu XQ. Application and challenges of stem cells in cardiovascular aging. Regen Ther 2024; 25:1-9. [PMID: 38108044 PMCID: PMC10724492 DOI: 10.1016/j.reth.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/17/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
With the rapid development of society and the economy, population aging has become a common challenge faced by many countries in the world today. Structural and functional changes in the cardiovascular system can occur with age, increasing the incidence and severity of cardiovascular diseases in older adults. Due to the limited regenerative capacity of myocardial cells, myocardial infarction and its resulting heart failure and congenital heart disease have become the number one killer of human health. At present, the treatment of cardiovascular diseases includes drug therapy and nondrug therapy. Nondrug therapy mainly includes minimally invasive interventional therapy, surgical diagnosis and treatment, and cell therapy. Long-term drug treatment may cause headache due to vasodilation, lower blood pressure, digestive system dysfunction and other side effects. Surgical treatment is traumatic, difficult to treat, and expensive. In recent years, stem cell therapy has exhibited broad application prospects in basic and clinical research on cardiovascular disease because of its plasticity, self-renewal and multidirectional differentiation potential. Therefore, this paper looks at stem cell therapy for diseases, reviews recent advances in the mechanism and clinical transformation of cardiovascular aging and related diseases in China, and briefly discusses the development trend and future prospects of cardiovascular aging research.
Collapse
Affiliation(s)
- He-Ling Yuan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Le Chang
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
| | - Wei-Wen Fan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Xin Liu
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qiang Li
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
- Kunming Medical University, Kunming, Yunnan 650500, China
| | - Chuan Tian
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
| | - Jing Zhao
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
| | - Zi-An Li
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
| | - Xing-Hua Pan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
| | - Xiang-Qing Zhu
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming 650032, Yunnan Province, China
| |
Collapse
|
17
|
Matta A, Ohlmann P, Nader V, Levai L, Kang R, Carrié D, Roncalli J. A review of the conservative versus invasive management of ischemic heart failure with reduced ejection fraction. Curr Probl Cardiol 2024; 49:102347. [PMID: 38103822 DOI: 10.1016/j.cpcardiol.2023.102347] [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: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Heart failure is increasing in terms of prevalence, morbidity, and mortality rates. Clinical trials and studies are focusing on heart failure as it is the destiny end-stage for several cardiovascular disorders. Recently, medical therapy has dramatically progressed with novel classes of medicines providing better quality of life and survival outcomes. However, heart failure remains a heavy impactful factor on societies and populations. Current guidelines from the American and European cardiac societies are not uniform with respect to the class and level of treatment recommendations for coronary artery disease patients with heart failure and reduced ejection fraction. The discrepancy among international recommendations, stemming from the lack of evidence from adequately powered randomized trials, challenges physicians in choosing the optimal strategy. Hybrid therapy including optimal medical therapy with revascularization strategies are commonly used for the management of ischemic heart failure. Coronary artery bypass graft (CABG) has proved its efficacy on improving long term outcome and prognosis while no large randomized clinical trials for percutaneous coronary intervention (PCI) are still available. Regardless of the lack of data and recommendations, the trends of performing PCI in ischemic heart failure prevailed over CABG whereas lesion complexity, chronic total occlusion and complete revascularization achievement are limiting factors. Lastly, regenerative medicine seems a promising approach for advanced heart failure enhancing cardiomyocytes proliferation, reverse remodeling, scar size reduction and cardiac function restoration.
Collapse
Affiliation(s)
- Anthony Matta
- Department of Cardiology, Civilian Hospital of Colmar, Colmar, France.
| | - Patrick Ohlmann
- Department of Cardiology, University Hospital of Strasbourg, Strasbourg, France
| | - Vanessa Nader
- Department of Cardiology, Civilian Hospital of Colmar, Colmar, France
| | - Laszlo Levai
- Department of Cardiology, Civilian Hospital of Colmar, Colmar, France
| | - Ryeonshi Kang
- Department of Cardiology, University Hospital of Toulouse, Toulouse, France
| | - Didier Carrié
- Department of Cardiology, University Hospital of Toulouse, Toulouse, France
| | - Jerome Roncalli
- Department of Cardiology, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
18
|
Eftekhar Z, Haybar H, Mohebbi A, Saki N. Cardiac Complications and COVID-19: A Review of Life-threatening Co-morbidities. Curr Cardiol Rev 2024; 20:1-12. [PMID: 38415433 PMCID: PMC11284692 DOI: 10.2174/011573403x279782240206091322] [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: 09/20/2023] [Revised: 12/23/2023] [Accepted: 01/19/2024] [Indexed: 02/29/2024] Open
Abstract
The novel 2019 coronavirus disease (COVID-19) was first reported in the last days of December 2019 in Wuhan, China. The presence of certain co-morbidities, including cardiovascular diseases (CVDs), are the basis for worse outcomes in patients with COVID-19. Relevant English-language literature was searched and retrieved from the Google Scholar search engine and PubMed database up to 2023 using COVID-19, SARS-CoV-2, Heart failure, Myocardial infarction, and Arrhythmia and Cardiac complication as keywords. Increased hemodynamic load, ischemia-related dysfunction, ventricular remodeling, excessive neurohumoral stimulation, abnormal myocyte calcium cycling, and excessive or insufficient extracellular matrix proliferation are associated with heart failure (HF) in COVID-19 patients. Inflammatory reaction due to the excessive release of inflammatory cytokines, leads to myocardial infarction (MI) in these patients. The virus can induce heart arrhythmia through cardiac complications, hypoxia, decreased heart hemodynamics, and remarkable inflammatory markers. Moreover, studies have linked cardiac complications in COVID-19 with poor outcomes, extended hospitalization time, and increased mortality rate. Patients with COVID-19 and CVDs are at higher mortality risk and they should be given high priority when receiving the treatment and intensive care during hospitalization.
Collapse
Affiliation(s)
- Zeinab Eftekhar
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Mohebbi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
19
|
He H, Yuan Y, Wu Y, Lu J, Yang X, Lu K, Liu A, Cao Z, Sun M, Yu M, Wang H. Exoskeleton Partial-Coated Stem Cells for Infarcted Myocardium Restoring. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2307169. [PMID: 37962473 DOI: 10.1002/adma.202307169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/30/2023] [Indexed: 11/15/2023]
Abstract
The integration of abiotic materials with live cells has emerged as an exciting strategy for the control of cellular functions. Exoskeletons consisting ofmetal-organic frameworks are generated to produce partial-coated bone marrow stem cells (BMSCs) to overcome low cell survival leading to disappointing effects for cell-based cardiac therapy. Partially coated exoskeletons can promote the survival of suspended BMSCs by integrating the support of exoskeletons and unimpaired cellular properties. In addition, partial exoskeletons exhibit protective effects against detrimental environmental conditions, including reactive oxygen species, pH changes, and osmotic pressure. The partial-coated cells exhibit increased intercellular adhesion forces to aggregate and adhere, promoting cell survival and preventing cell escape during cell therapy. The exoskeletons interact with cell surface receptors integrin α5β1, leading to augmented biological functions with profitable gene expression alteration, such as Vegfa, Cxcl12, and Adm. The partial-coated BMSCs display enhanced cell retention in infarcted myocardium through non-invasive intravenous injections. The repair of myocardial infarction has been achieved with improved cardiac function, myocardial angiogenesis, proliferation, and inhibition of cell apoptosis. This discovery advances the elucidation of potential molecular and cellular mechanisms for cell-exoskeleton interactions and benefits the rational design and manufacture of next-generation nanobiohybrids.
Collapse
Affiliation(s)
- Huihui He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Yuan Yuan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, 310058, China
| | - Yunhong Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jingyi Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Xiaofu Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Kejie Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - An Liu
- Department of Orthopaedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310009, China
| | - Zelin Cao
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Miao Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
20
|
Mukkala AN, Jerkic M, Khan Z, Szaszi K, Kapus A, Rotstein O. Therapeutic Effects of Mesenchymal Stromal Cells Require Mitochondrial Transfer and Quality Control. Int J Mol Sci 2023; 24:15788. [PMID: 37958771 PMCID: PMC10647450 DOI: 10.3390/ijms242115788] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Due to their beneficial effects in an array of diseases, Mesenchymal Stromal Cells (MSCs) have been the focus of intense preclinical research and clinical implementation for decades. MSCs have multilineage differentiation capacity, support hematopoiesis, secrete pro-regenerative factors and exert immunoregulatory functions promoting homeostasis and the resolution of injury/inflammation. The main effects of MSCs include modulation of immune cells (macrophages, neutrophils, and lymphocytes), secretion of antimicrobial peptides, and transfer of mitochondria (Mt) to injured cells. These actions can be enhanced by priming (i.e., licensing) MSCs prior to exposure to deleterious microenvironments. Preclinical evidence suggests that MSCs can exert therapeutic effects in a variety of pathological states, including cardiac, respiratory, hepatic, renal, and neurological diseases. One of the key emerging beneficial actions of MSCs is the improvement of mitochondrial functions in the injured tissues by enhancing mitochondrial quality control (MQC). Recent advances in the understanding of cellular MQC, including mitochondrial biogenesis, mitophagy, fission, and fusion, helped uncover how MSCs enhance these processes. Specifically, MSCs have been suggested to regulate peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC1α)-dependent biogenesis, Parkin-dependent mitophagy, and Mitofusins (Mfn1/2) or Dynamin Related Protein-1 (Drp1)-mediated fission/fusion. In addition, previous studies also verified mitochondrial transfer from MSCs through tunneling nanotubes and via microvesicular transport. Combined, these effects improve mitochondrial functions, thereby contributing to the resolution of injury and inflammation. Thus, uncovering how MSCs affect MQC opens new therapeutic avenues for organ injury, and the transplantation of MSC-derived mitochondria to injured tissues might represent an attractive new therapeutic approach.
Collapse
Affiliation(s)
- Avinash Naraiah Mukkala
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mirjana Jerkic
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
| | - Zahra Khan
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Katalin Szaszi
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Andras Kapus
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ori Rotstein
- Unity Health Toronto, The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, University of Toronto, Toronto, ON M5B 1T8, Canada; (A.N.M.); (Z.K.); (K.S.); (A.K.); (O.R.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
21
|
Schubart JR, Zare A, Fernandez-de-Castro RM, Figueroa HR, Sarel I, Tuchman K, Esposito K, Henderson FC, von Schwarz E. Safety and outcomes analysis: transcatheter implantation of autologous angiogenic cell precursors for the treatment of cardiomyopathy. Stem Cell Res Ther 2023; 14:308. [PMID: 37880753 PMCID: PMC10601268 DOI: 10.1186/s13287-023-03539-6] [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: 06/16/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Stem cell transplantation is an emerging therapy for severe cardiomyopathy, proffering stem cell recruitment, anti-apoptosis, and proangiogenic capabilities. Angiogenic cell precursors (ACP-01) are autologous, lineage-specific, cells derived from a multipotent progenitor cell population, with strong potential to effectively engraft, form blood vessels, and support tissue survival and regeneration. METHODS This IRB approved outcome analysis reports upon 74 consecutive patients who failed medical management for severe cardiomyopathy, and were selected to undergo transcatheter intramyocardial or intracoronary implantation of ACP-01. Serious adverse events (SAEs) were reported. Cell analysis was conducted for each treatment. The left ventricular ejection fraction (LVEF) was measured by multi-gated acquisition scan (MUGA) or echocardiogram at 4 months ± 1.9 months and 12 months ± 5.5 months. Patients reported quality of life statements at 6 months (± 5.6 months). RESULTS Fifty-four of 74 patients met requirements for inclusion (48 males and five females; age 68.1 ± 11.3 years). The mean treatment cell number of 57 × 106 ACP-01 included 7.7 × 106 CD34 + and 21 × 106 CD31 + cells with 97.6% viability. SAEs included one death (previously unrecognized silent MI), ventricular tachycardia (n = 2) requiring cardioversion, and respiratory infection (n = 2). LVEF in the ischemic subgroup (n = 41) improved by 4.7% ± 9.7 from pre-procedure to the first follow-up (4 months ± 1.9 months) (p < 0.004) and by 7.2% ± 10.9 at final follow-up (n = 25) at average 12 months (p < 0.004). The non-ischemic dilated cardiomyopathy subgroup (n = 8) improved by 7.5% ± 6.0 at the first follow-up (p < 0.017) and by 12.2% ± 6.4 at final follow-up (p < 0.003, n = 6). Overall improvement in LVEF from pre-procedure to post-procedure was significant (Fisher's exact test p < 0.004). LVEF improvement was most marked in the patients with the most severe cardiomyopathy (LVEF < 20%) improving from a mean 14.6% ± 3.4% pre-procedurally to 28.4% ± 8% at final follow-up. Quality of life statements reflected improvement in 33/50 (66%), no change in 14/50 (28%), and worse in 3/50 (6%). CONCLUSION Transcatheter implantation of ACP-01 for cardiomyopathy is safe and improves LVEF in the setting of ischemic and non-ischemic cardiomyopathy. The results warrant further investigation in a prospective, blinded, and controlled clinical study. TRIAL REGISTRATION IRB from Genetic Alliance #APC01-001, approval date July 25, 2022. Cardiomyopathy is common and associated with high mortality. Stem cell transplantation is an emerging therapy. Angiogenic cell precursors (ACP-01) are lineage-specific endothelial progenitors, with strong potential for migration, engraftment, angiogenesis, and support of tissue survival and regeneration. A retrospective outcomes analysis of 53 patients with ischemic and non-ischemic dilated cardiomyopathy undergoing transcatheter implantation of ACP-01 demonstrated improvements in the left ventricular ejection fraction of 7.2% ± 10.9 (p < 0.004) and 12.2% ± 6.4, respectively, at 12 months (± 5) follow-up. Quality of life statements reflected improvement in 33/50 (66%) patients.
Collapse
Affiliation(s)
- Jane R Schubart
- Penn State College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Amirhossein Zare
- Northern Ontario School of Medicine, Ontario, CA, USA
- Hemostemix Inc, Calgary, CA, Canada
| | | | | | | | - Kelly Tuchman
- The Metropolitan Neurosurgery Group, LLC, 1010 Wayne Ave Suite 420, Silver Spring, MD, 20910, USA.
| | - Kaitlyn Esposito
- The Bobby Jones Chiari Syringomyelia Foundation, New York, NY, USA
| | - Fraser C Henderson
- The Metropolitan Neurosurgery Group, LLC, 1010 Wayne Ave Suite 420, Silver Spring, MD, 20910, USA.
- Department Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.
- Hemostemix Inc, Calgary, CA, Canada.
| | - Ernst von Schwarz
- School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Cedars Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
22
|
Carbone RG, Negrini S, Murdaca G, Fontana V, Puppo F. Stem cells treatment in chronic ischemic heart disease: a narrative review. AMERICAN JOURNAL OF STEM CELLS 2023; 12:65-72. [PMID: 38021453 PMCID: PMC10658134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023]
Abstract
Chronic ischemic heart disease remains a major cause of morbidity and mortality worldwide. Several trials have been performed to evaluate benefit of stem cells transplantation to restore cardiac function in short- and long-term period after myocardial infarction. This narrative review analyzes 24 clinical trials between 2005 and 2023 comprising 1824 patients with chronic heart disease without heart failure. Percent increase in left ventricular ejection fraction (LVEF) and decrease in New York Heart Association (NYHA) class at 6/12 months after stem cells transplantation are reported. Thirteen trials showed a statistically significant percent LVEF increase between 4% to 19% at 6/12 months after stem cells transplantation (p values from 0.05 to 0.0001). No significant differences in LVEF were observed between patients who underwent intracoronary or intramyocardial transplantation. NYHA class decrease from severe to mild/moderate was demonstrated in 10 trials reporting a significant LVEF increase. Patients transplanted with bone marrow and peripheral blood CD133+ stem cells showed a doubling of percentage LVEF increase in comparison to patients transplanted with CD133- cells. This narrative review reports the conflicting results on this topic. Multicenter randomized clinical trials should be performed to define the efficacy of stem cells transplantation in chronic ischemic heart disease.
Collapse
Affiliation(s)
| | - Simone Negrini
- Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of GenoaGenoa, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology Unit, IRCCS San Martino HospitalGenoa, Italy
| | - Francesco Puppo
- Department of Internal Medicine, University of GenoaGenoa, Italy
| |
Collapse
|
23
|
Muneer R, Qazi REM, Fatima A, Ahmad W, Salim A, Dini L, Khan I. Wnt signaling pathway inhibitor promotes mesenchymal stem cells differentiation into cardiac progenitor cells in vitro and improves cardiomyopathy in vivo. World J Stem Cells 2023; 15:821-841. [PMID: 37700819 PMCID: PMC10494566 DOI: 10.4252/wjsc.v15.i8.821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/31/2023] [Accepted: 07/03/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Cardiovascular diseases particularly myocardial infarction (MI) are the leading cause of mortality and morbidity around the globe. As cardiac tissue possesses very limited regeneration potential, therefore use of a potent small molecule, inhibitor Wnt production-4 (IWP-4) for stem cell differentiation into cardiomyocytes could be a promising approach for cardiac regeneration. Wnt pathway inhibitors may help stem cells in their fate determination towards cardiomyogenic lineage and provide better homing and survival of cells in vivo. Mesenchymal stem cells (MSCs) derived from the human umbilical cord have the potential to regenerate cardiac tissue, as they are easy to isolate and possess multilineage differentiation capability. IWP-4 may promote the differentiation of MSCs into the cardiac lineage. AIM To evaluate the cardiac differentiation ability of IWP-4 and its subsequent in vivo effects. METHODS Umbilical cord tissue of human origin was utilized to isolate the MSCs which were characterized by their morphology, immunophenotyping of surface markers specific to MSCs, as well as by tri-lineage differentiation capability. Cytotoxicity analysis was performed to identify the optimal concentration of IWP-4. MSCs were treated with 5 μM IWP-4 at two different time intervals. Differentiation of MSCs into cardiomyocytes was evaluated at DNA and protein levels. The MI rat model was developed. IWP-4 treated as well as untreated MSCs were implanted in the MI model, then the cardiac function was analyzed via echocardiography. MSCs were labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI) dye for tracking, while the regeneration of infarcted myocardium was examined by histology and immunohistochemistry. RESULTS MSCs were isolated and characterized. Cytotoxicity analysis showed that IWP-4 was non-cytotoxic at 5 μM concentration. Cardiac specific gene and protein expression analyses exhibited more remarkable results in fourteen days treated group that was eventually selected for in vivo transplantation. Cardiac function was restored in the IWP-4 treated group in comparison to the MI group. Immunohistochemical analysis confirmed the homing of pre-differentiated MSCs that were labeled with DiI cell labeling dye. Histological analysis confirmed the significant reduction in fibrotic area, and improved left ventricular wall thickness in IWP-4 treated MSC group. CONCLUSION Treatment of MSCs with IWP-4 inhibits Wnt pathway and promotes cardiac differentiation. These pre-conditioned MSCs transplanted in vivo improved cardiac function by cell homing, survival, and differentiation at the infarcted region, increased left ventricular wall thickness, and reduced infarct size.
Collapse
Affiliation(s)
- Rabbia Muneer
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Rida-E-Maria Qazi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Abiha Fatima
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Waqas Ahmad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Luciana Dini
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, Rome 00185, Italy
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan.
| |
Collapse
|
24
|
Ando Y, Chang FC, James M, Zhou Y, Zhang M. Chitosan Scaffolds as Microcarriers for Dynamic Culture of Human Neural Stem Cells. Pharmaceutics 2023; 15:1957. [PMID: 37514142 PMCID: PMC10384976 DOI: 10.3390/pharmaceutics15071957] [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: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Human neural stem cells (hNSCs) possess remarkable potential for regenerative medicine in the treatment of presently incurable diseases. However, a key challenge lies in producing sufficient quantities of hNSCs, which is necessary for effective treatment. Dynamic culture systems are recognized as a powerful approach to producing large quantities of hNSCs required, where microcarriers play a critical role in supporting cell expansion. Nevertheless, the currently available microcarriers have limitations, including a lack of appropriate surface chemistry to promote cell adhesion, inadequate mechanical properties to protect cells from dynamic forces, and poor suitability for mass production. Here, we present the development of three-dimensional (3D) chitosan scaffolds as microcarriers for hNSC expansion under defined conditions in bioreactors. We demonstrate that chitosan scaffolds with a concentration of 4 wt% (4CS scaffolds) exhibit desirable microstructural characteristics and mechanical properties suited for hNSC expansion. Furthermore, they could also withstand degradation in dynamic conditions. The 4CS scaffold condition yields optimal metabolic activity, cell adhesion, and protein expression, enabling sustained hNSC expansion for up to three weeks in a dynamic culture. Our study introduces an effective microcarrier approach for prolonged expansion of hNSCs, which has the potential for mass production in a three-dimensional setting.
Collapse
Affiliation(s)
- Yoshiki Ando
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu 520-2362, Shiga, Japan
| | - Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Matthew James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
25
|
Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS, Bragança J, Calado SM. Human Stem Cells for Cardiac Disease Modeling and Preclinical and Clinical Applications—Are We on the Road to Success? Cells 2023; 12:1727. [DOI: https:/doi.org/10.3390/cells12131727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are pointed out by the World Health Organization (WHO) as the leading cause of death, contributing to a significant and growing global health and economic burden. Despite advancements in clinical approaches, there is a critical need for innovative cardiovascular treatments to improve patient outcomes. Therapies based on adult stem cells (ASCs) and embryonic stem cells (ESCs) have emerged as promising strategies to regenerate damaged cardiac tissue and restore cardiac function. Moreover, the generation of human induced pluripotent stem cells (iPSCs) from somatic cells has opened new avenues for disease modeling, drug discovery, and regenerative medicine applications, with fewer ethical concerns than those associated with ESCs. Herein, we provide a state-of-the-art review on the application of human pluripotent stem cells in CVD research and clinics. We describe the types and sources of stem cells that have been tested in preclinical and clinical trials for the treatment of CVDs as well as the applications of pluripotent stem-cell-derived in vitro systems to mimic disease phenotypes. How human stem-cell-based in vitro systems can overcome the limitations of current toxicological studies is also discussed. Finally, the current state of clinical trials involving stem-cell-based approaches to treat CVDs are presented, and the strengths and weaknesses are critically discussed to assess whether researchers and clinicians are getting closer to success.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anita Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- School of Health, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bárbara M. Silva
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
26
|
Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS, Bragança J, Calado SM. Human Stem Cells for Cardiac Disease Modeling and Preclinical and Clinical Applications-Are We on the Road to Success? Cells 2023; 12:1727. [PMID: 37443761 PMCID: PMC10341347 DOI: 10.3390/cells12131727] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Cardiovascular diseases (CVDs) are pointed out by the World Health Organization (WHO) as the leading cause of death, contributing to a significant and growing global health and economic burden. Despite advancements in clinical approaches, there is a critical need for innovative cardiovascular treatments to improve patient outcomes. Therapies based on adult stem cells (ASCs) and embryonic stem cells (ESCs) have emerged as promising strategies to regenerate damaged cardiac tissue and restore cardiac function. Moreover, the generation of human induced pluripotent stem cells (iPSCs) from somatic cells has opened new avenues for disease modeling, drug discovery, and regenerative medicine applications, with fewer ethical concerns than those associated with ESCs. Herein, we provide a state-of-the-art review on the application of human pluripotent stem cells in CVD research and clinics. We describe the types and sources of stem cells that have been tested in preclinical and clinical trials for the treatment of CVDs as well as the applications of pluripotent stem-cell-derived in vitro systems to mimic disease phenotypes. How human stem-cell-based in vitro systems can overcome the limitations of current toxicological studies is also discussed. Finally, the current state of clinical trials involving stem-cell-based approaches to treat CVDs are presented, and the strengths and weaknesses are critically discussed to assess whether researchers and clinicians are getting closer to success.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Anita Ferreira
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- School of Health, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Bárbara M. Silva
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (A.F.); (M.T.F.); (B.M.S.); (F.E.); (H.S.L.); (J.B.)
- Algarve Biomedical Center (ABC), Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, Universidade do Algarve—Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
27
|
Phinney DG, Hwa Lee R, Boregowda SV. Revisiting the Mesenchymal "Stem vs. Stromal" Cell Dichotomy and Its Implications for Development of Improved Potency Metrics. Stem Cells 2023; 41:444-452. [PMID: 36891977 PMCID: PMC10183967 DOI: 10.1093/stmcls/sxad019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 03/10/2023]
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapies have been evaluated in over 1500 human clinical trials for a diverse array of disease indication, but outcomes remain unpredictable due to knowledge gaps in the quality attributes that confer therapeutic potency onto cells and their mode of action in vivo. Based on accumulated evidence from pre-clinical models, MSCs exert therapeutic effects by repressing inflammatory and immune-mediated response via paracrine action following reprogramming by the host injury microenvironment, and by polarization of tissue resident macrophages following phagocytosis to an alternatively activated (M2) state. An important tenet of this existing paradigm is that well-established stem/progenitor functions of MSCs are independent of paracrine function and dispensable for their anti-inflammatory and immune suppressive functions. Herein, we review evidence that stem/progenitor and paracrine functions of MSCs are mechanistically linked and organized hierarchically and describe how this link may be exploited to develop metrics that predict MSC potency across a spectrum of activities and regenerative medicine applications.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA
| | - Siddaraju V Boregowda
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| |
Collapse
|
28
|
Todorova VB, Baxan N, Delahaye M, Harding SE, Rankin SM. Drug-based mobilisation of mesenchymal stem/stromal cells improves cardiac function post myocardial infarction. Dis Model Mech 2023; 16:dmm049630. [PMID: 36263604 PMCID: PMC10655717 DOI: 10.1242/dmm.049630] [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: 05/04/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
Abstract
There is an unmet need for treatments that prevent the progressive cardiac dysfunction following myocardial infarction. Mesenchymal stem/stromal cells (MSCs) are under investigation for cardiac repair; however, culture expansion prior to transplantation is hindering their homing and reparative abilities. Pharmacological mobilisation could be an alternative to MSC transplantation. Here, we report that endogenous MSCs mobilise into the circulation at day 5 post myocardial infarction in male Lewis rats. This mobilisation can be significantly increased by using a combination of the FDA-approved drugs mirabegron (β3-adrenoceptor agonist) and AMD3100 (CXCR4 antagonist). Blinded cardiac magnetic resonance imaging analysis showed the treated group to have increased left ventricular ejection fraction and decreased end systolic volume at 5 weeks post myocardial infarction. The mobilised group had a significant decrease in plasma IL-6 and TNF-α levels, a decrease in interstitial fibrosis, and an increase in the border zone blood vessel density. Conditioned medium from blood-derived MSCs supported angiogenesis in vitro, as shown by tube formation and wound healing assays. Our data suggest a novel pharmacological strategy that enhances myocardial infarction-induced MSC mobilisation and improves cardiac function after myocardial infarction.
Collapse
Affiliation(s)
- Veneta B. Todorova
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Nicoleta Baxan
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Matthew Delahaye
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Sian E. Harding
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Sara M. Rankin
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| |
Collapse
|
29
|
Zheng PF, Liu F, Zheng ZF, Pan HW, Liu ZY. Identification MNS1, FRZB, OGN, LUM, SERP1NA3 and FCN3 as the potential immune-related key genes involved in ischaemic cardiomyopathy by random forest and nomogram. Aging (Albany NY) 2023; 15:1475-1495. [PMID: 36863704 PMCID: PMC10042686 DOI: 10.18632/aging.204547] [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: 11/27/2022] [Accepted: 02/11/2023] [Indexed: 03/04/2023]
Abstract
The immune molecular mechanisms involved in ischaemic cardiomyopathy (ICM) have not been fully elucidated. The current study aimed to elucidate the immune cell infiltration pattern of the ICM and identify key immune-related genes that participate in the pathologic process of the ICM. The differentially expressed genes (DEGs) were identified from two datasets (GSE42955 combined with GSE57338) and the top 8 key DEGs related to ICM were screened using random forest and used to construct the nomogram model. Moreover, the "CIBERSORT" software package was used to determine the proportion of infiltrating immune cells in the ICM. A total of 39 DEGs (18 upregulated and 21 downregulated) were identified in the current study. Four upregulated DEGs, including MNS1, FRZB, OGN, and LUM, and four downregulated DEGs, SERP1NA3, RNASE2, FCN3 and SLCO4A1, were identified by the random forest model. The nomogram constructed based on the above 8 key genes suggested a diagnostic value of up to 99% to distinguish the ICM from healthy participants. Meanwhile, most of the key DEGs presented prominent interactions with immune cell infiltrates. The RT-qPCR results suggested that the expression levels of MNS1, FRZB, OGN, LUM, SERP1NA3 and FCN3 between the ICM and control groups were consistent with the bioinformatic analysis results. These results suggested that immune cell infiltration plays a critical role in the occurrence and progression of ICM. Several key immune-related genes, including the MNS1, FRZB, OGN, LUM, SERP1NA3 and FCN3 genes, are expected to be reliable serum markers for the diagnosis of ICM and potential molecular targets for ICM immunotherapy.
Collapse
Affiliation(s)
- Peng-Fei Zheng
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Fen Liu
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Furong, Changsha 410000, Hunan, China
| | - Zhao-Fen Zheng
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Hong-Wei Pan
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| | - Zheng-Yu Liu
- Cardiology Department, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
- Clinical Research Center for Heart Failure in Hunan Province, Furong, Changsha 410000, Hunan, China
- Institute of Cardiovascular Epidemiology, Hunan Provincial People’s Hospital, Furong, Changsha 410000, Hunan, China
| |
Collapse
|
30
|
Dezhakam E, Khalilzadeh B, Mahdipour M, Isildak I, Yousefi H, Ahmadi M, Naseri A, Rahbarghazi R. Electrochemical biosensors in exosome analysis; a short journey to the present and future trends in early-stage evaluation of cancers. Biosens Bioelectron 2023; 222:114980. [PMID: 36521207 DOI: 10.1016/j.bios.2022.114980] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/15/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment consists of a multiplicity of cells such as cancer cells, fibroblasts, endothelial cells, and immune cells within the specific parenchyma. It has been indicated that cancer cells can educate other cells within the tumor niche in a paracrine manner by the release of nano-sized extracellular vesicles namely exosomes (Exo), resulting in accelerated tumor mass growth. It is suggested that exosomal cargo with remarkable information can reflect any changes in metabolic and proteomic profiles in parent tumor cells. Therefore, exosomes can be touted as prognostic, diagnostic, and therapeutic elements with specific biomarkers in patients with different tumor types. Despite the advantages, conventional exosome separation and purification protocols are time-consuming and laborious with low abnormal morphology and purity rate. During the last decades, biosensor-based modalities, as emerging instruments, have been used to detect and analyze Exo in biofluids. Due to suitable specificity, sensitivity, and real-time readout, biosensors became promising approaches for the analysis of Exo in in vitro and in vivo settings. The inherent advantages and superiority of electrochemical biosensors in the determination of tumor grade based on exosomal cargo and profile were also debated. Present and future challenges were also discussed related to the application of electrochemical biosensors in the clinical setting. In this review, the early detection of several cancer types associated with ovaries, breast, brain, colon, lungs, T and B lymphocytes, liver and rare types of cancers were debated in association with released exosomes.
Collapse
Affiliation(s)
- Ehsan Dezhakam
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ibrahim Isildak
- Department of Bioengineering, Faculty of Chemistry-Metallurgy, Yildiz Technical University, 34220, Istanbul, Turkey
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdolhossein Naseri
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Vignais ML, Levoux J, Sicard P, Khattar K, Lozza C, Gervais M, Mezhoud S, Nakhle J, Relaix F, Agbulut O, Fauconnier J, Rodriguez AM. Transfer of Cardiac Mitochondria Improves the Therapeutic Efficacy of Mesenchymal Stem Cells in a Preclinical Model of Ischemic Heart Disease. Cells 2023; 12:cells12040582. [PMID: 36831249 PMCID: PMC9953768 DOI: 10.3390/cells12040582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND The use of mesenchymal stem cells (MSCs) appears to be a promising therapeutic approach for cardiac repair after myocardial infarction. However, clinical trials have revealed the need to improve their therapeutic efficacy. Recent evidence demonstrated that mitochondria undergo spontaneous transfer from damaged cells to MSCs, resulting in the activation of the cytoprotective and pro-angiogenic functions of recipient MSCs. Based on these observations, we investigated whether the preconditioning of MSCs with mitochondria could optimize their therapeutic potential for ischemic heart disease. METHODS Human MSCs were exposed to mitochondria isolated from human fetal cardiomyocytes. After 24 h, the effects of mitochondria preconditioning on the MSCs' function were analyzed both in vitro and in vivo. RESULTS We found that cardiac mitochondria-preconditioning improved the proliferation and repair properties of MSCs in vitro. Mechanistically, cardiac mitochondria mediate their stimulatory effects through the production of reactive oxygen species, which trigger their own degradation in recipient MSCs. These effects were further confirmed in vivo, as the mitochondria preconditioning of MSCs potentiated their therapeutic efficacy on cardiac function following their engraftment into infarcted mouse hearts. CONCLUSIONS The preconditioning of MSCs with the artificial transfer of cardiac mitochondria appears to be promising strategy to improve the efficacy of MSC-based cell therapy in ischemic heart disease.
Collapse
Affiliation(s)
- Marie-Luce Vignais
- Institut de Génomique Fonctionnelle, University Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Jennyfer Levoux
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM U1164, Biological Adaptation and Ageing, 75005 Paris, France
| | - Pierre Sicard
- PhyMedExp, Inserm, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Khattar Khattar
- Institut de Génomique Fonctionnelle, University Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Catherine Lozza
- PhyMedExp, Inserm, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Marianne Gervais
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Safia Mezhoud
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Jean Nakhle
- Institut de Génomique Fonctionnelle, University Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Frederic Relaix
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
- APHP, Hôpitaux Universitaires Henri Mondor & Centre de Référence des Maladies Neuromusculaires GNMH, 94000 Créteil, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM U1164, Biological Adaptation and Ageing, 75005 Paris, France
| | - Jeremy Fauconnier
- PhyMedExp, Inserm, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Anne-Marie Rodriguez
- Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM U1164, Biological Adaptation and Ageing, 75005 Paris, France
- APHP, Hôpitaux Universitaires Henri Mondor & Centre de Référence des Maladies Neuromusculaires GNMH, 94000 Créteil, France
- Correspondence:
| |
Collapse
|
32
|
Ghoreishi AS, Iranmanesh E, Rastegarpouyani H, Mokhtarian S, Poshtchaman Z, Javadi ZS, Khoshdel A. Better isolation, proliferation and differentiation of human adipose-derived mesenchymal stem cells using human serum. Eur J Transl Myol 2023; 33. [PMID: 36714911 PMCID: PMC10141746 DOI: 10.4081/ejtm.2023.10834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/30/2022] [Indexed: 01/28/2023] Open
Abstract
Mesenchymal stem cells have many applications in medicine. Attention to the proliferation and differentiation of stem cell differentiation is an important issue. The aim of this study was to investigate the possibility of optimal isolation, proliferation, and differentiation of adipose tissue-derived mesenchymal stem cells (ADSCs) using human serum. Human serum (HS) was obtained from the venous blood of eight healthy individuals. The rate of proliferation and differentiation of ADSCs and expression of surface markers was assessed by flow cytometry. Bone differentiation was assessed using Alizarin Red staining. Data were analyzed using statistical software. Over time, HS showed more proliferation than fetal bovine serum (FBS) -enriched cells (p <0.05). Differentiation of ADSCs cells ls in HS-enriched medium is faster and more pronounced than differentiation in the control group. The expression of surface markers in the medium containing HS was the same as the medium containing FBS where the expression levels of CD105 and CD95 were found to be positive and the expression of CD34 and CD45 was negative. Due to the better proliferation of adipose tissue-derived mesenchymal cells in the medium containing HS than FBS, it is suggested that human serum be used in future clinical studies. Also, HS is healthier, safer, more accessible, and more affordable than FBS.
Collapse
Affiliation(s)
- Atena Sadat Ghoreishi
- Department of Clinical Biochemistry, Faculty of Para-Medicine, Jiroft University of Medical Sciences, Jiroft.
| | - Ehsan Iranmanesh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman.
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL, USA Institute for Molecular Biophysics, Florida State University, Tallahassee, FL.
| | - Sogand Mokhtarian
- Department of Cell and Molecular Biology, Islamic Azad University of Shahr-e-Qods, Tehran.
| | - Zahra Poshtchaman
- MSc of Critical care Nursing, Department of Nursing, Esfarayen Faculty of Medical Sciences, Esfarayen.
| | - Zeinab Sadat Javadi
- Yazd hospital Mehrab Shohada, Shahid Sadoughi University of Medical Sciences, Yazd.
| | - Alireza Khoshdel
- Department of Clinical Biochemistry, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan.
| |
Collapse
|
33
|
Maeda S, Kawamura T, Chida D, Shimamura K, Toda K, Harada A, Sawa Y, Miyagawa S. Notch Signaling-Modified Mesenchymal Stem Cell Patch Improves Left Ventricular Function via Arteriogenesis Induction in a Rat Myocardial Infarction Model. Cell Transplant 2023; 32:9636897231154580. [PMID: 36946544 PMCID: PMC10037722 DOI: 10.1177/09636897231154580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
For ischemic cardiomyopathy (ICM) with limited therapeutic options, the induction of arteriogenesis has the potential to improve cardiac function through major restoration of blood flow. We hypothesized that transplantation of a Notch signaling-modified mesenchymal stem cell (SB623 cell) patch would induce angiogenesis and arteriogenesis in ischemic lesions, leading to improvement of left ventricular (LV) function in a rat ICM model. Two weeks after the induction of ischemia, SB623 cell patch transplantation into ICM rats (SB group, n = 10) or a sham operation (no-treatment group, n = 10) was performed. The LV ejection fraction was significantly improved at 6 weeks after SB623 cell patch transplantation (P < 0.001). Histological findings revealed that the number of von Willebrand factor (vWF)-positive capillary vessels (P < 0.01) and alpha smooth muscle actin (αSMA)- and vWF-positive arterioles with a diameter greater than 20 µm (P = 0.002) was significantly increased in the SB group, suggesting the induction of angiogenesis and arteriogenesis. Moreover, rat cardiomyocytes treated with SB623 cell patch transplantation showed upregulation of ephrin-B2 (P = 0.03) and EphB4 (P = 0.01) gene expression, indicating arteriogenesis induction. In conclusion, SB623 cell patch transplantation improved LV function by inducing angiogenesis and arteriogenesis in a rat ICM model.
Collapse
Affiliation(s)
- Shusaku Maeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Kazuo Shimamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koichi Toda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
34
|
Qazi REM, Khan I, Haneef K, Malick TS, Naeem N, Ahmad W, Salim A, Mohsin S. Combination of mesenchymal stem cells and three-dimensional collagen scaffold preserves ventricular remodeling in rat myocardial infarction model. World J Stem Cells 2022; 14:633-657. [PMID: 36157910 PMCID: PMC9453269 DOI: 10.4252/wjsc.v14.i8.633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/09/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiovascular diseases are the major cause of mortality worldwide. Regeneration of the damaged myocardium remains a challenge due to mechanical constraints and limited healing ability of the adult heart tissue. Cardiac tissue engineering using biomaterial scaffolds combined with stem cells and bioactive molecules could be a highly promising approach for cardiac repair. Use of biomaterials can provide suitable microenvironment to the cells and can solve cell engraftment problems associated with cell transplantation alone. Mesenchymal stem cells (MSCs) are potential candidates in cardiac tissue engineering because of their multilineage differentiation potential and ease of isolation. Use of DNA methyl transferase inhibitor, such as zebularine, in combination with three-dimensional (3D) scaffold can promote efficient MSC differentiation into cardiac lineage, as epigenetic modifications play a fundamental role in determining cell fate and lineage specific gene expression. AIM To investigate the role of collagen scaffold and zebularine in the differentiation of rat bone marrow (BM)-MSCs and their subsequent in vivo effects. METHODS MSCs were isolated from rat BM and characterized morphologically, immunophenotypically and by multilineage differentiation potential. MSCs were seeded in collagen scaffold and treated with 3 μmol/L zebularine in three different ways. Cytotoxicity analysis was done and cardiac differentiation was analyzed at the gene and protein levels. Treated and untreated MSC-seeded scaffolds were transplanted in the rat myocardial infarction (MI) model and cardiac function was assessed by echocardiography. Cell tracking was performed by DiI dye labeling, while regeneration and neovascularization were evaluated by histological and immunohistochemical analysis, res pectively. RESULTS MSCs were successfully isolated and seeded in collagen scaffold. Cytotoxicity analysis revealed that zebularine was not cytotoxic in any of the treatment groups. Cardiac differentiation analysis showed more pronounced results in the type 3 treatment group which was subsequently chosen for the transplantation in the in vivo MI model. Significant improvement in cardiac function was observed in the zebularine treated MSC-seeded scaffold group as compared to the MI control. Histological analysis also showed reduction in fibrotic scar, improvement in left ventricular wall thickness and preservation of ventricular remodeling in the zebularine treated MSC-seeded scaffold group. Immunohistochemical analysis revealed significant expression of cardiac proteins in DiI labeled transplanted cells and a significant increase in the number of blood vessels in the zebularine treated MSC-seeded collagen scaffold transplanted group. CONCLUSION Combination of 3D collagen scaffold and zebularine treatment enhances cardiac differentiation potential of MSCs, improves cell engraftment at the infarcted region, reduces infarct size and improves cardiac function.
Collapse
Affiliation(s)
- Rida-E-Maria Qazi
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Irfan Khan
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Kanwal Haneef
- Dr.Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Tuba Shakil Malick
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Nadia Naeem
- Dow Research Institute of Biotechnology and Biomedical Sciences (DRIBBS), Dow University of Health and Sciences, Ojha Campus, Karachi 74200, Sindh, Pakistan
| | - Waqas Ahmad
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Asmat Salim
- Stem Cell Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, ICCBS, University of Karachi, Karachi 75270, Sindh, Pakistan.
| | - Sadia Mohsin
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States
| |
Collapse
|
35
|
Liao R, Li Z, Wang Q, Lin H, Sun H. Revascularization of chronic total occlusion coronary artery and cardiac regeneration. Front Cardiovasc Med 2022; 9:940808. [PMID: 36093131 PMCID: PMC9455703 DOI: 10.3389/fcvm.2022.940808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Coronary chronic total occlusion (CTO) contributes to the progression of heart failure in patients with ischemic cardiomyopathy. Randomized controlled trials demonstrated that percutaneous coronary intervention (PCI) for CTO significantly improves angina symptoms and quality of life but fails to reduce clinical events compared with optimal medical therapy. Even so, intervening physicians strongly support CTO-PCI. Cardiac regeneration therapy after CTO-PCI should be a promising approach to improving the prognosis of ischemic cardiomyopathy. However, the relationship between CTO revascularization and cardiac regeneration has rarely been studied, and experimental studies on cardiac regeneration usually employ rodent models with permanent ligation of the coronary artery rather than reopening of the occlusive artery. Limited early-stage clinical trials demonstrated that cell therapy for cardiac regeneration in ischemic cardiomyopathy reduces scar size, reverses cardiac remodeling, and promotes angiogenesis. This review focuses on the status quo of CTO-PCI in ischemic cardiomyopathy and the clinical prospect of cardiac regeneration in this setting.
Collapse
Affiliation(s)
- Ruoxi Liao
- Department of Clinical Medicine, Dalian Medical University, Dalian, China
| | - Zhihong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiancheng Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hairuo Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Hairuo Lin, ,
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Huijun Sun,
| |
Collapse
|
36
|
Yedavilli S, Singh AD, Singh D, Samal R. Nano-Messengers of the Heart: Promising Theranostic Candidates for Cardiovascular Maladies. Front Physiol 2022; 13:895322. [PMID: 35899033 PMCID: PMC9313536 DOI: 10.3389/fphys.2022.895322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Till date, cardiovascular diseases remain a leading cause of morbidity and mortality across the globe. Several commonly used treatment methods are unable to offer safety from future complications and longevity to the patients. Therefore, better and more effective treatment measures are needed. A potential cutting-edge technology comprises stem cell-derived exosomes. These nanobodies secreted by cells are intended to transfer molecular cargo to other cells for the establishment of intercellular communication and homeostasis. They carry DNA, RNA, lipids, and proteins; many of these molecules are of diagnostic and therapeutic potential. Several stem cell exosomal derivatives have been found to mimic the cardioprotective attributes of their parent stem cells, thus holding the potential to act analogous to stem cell therapies. Their translational value remains high as they have minimal immunogenicity, toxicity, and teratogenicity. The current review highlights the potential of various stem cell exosomes in cardiac repair, emphasizing the recent advancements made in the development of cell-free therapeutics, particularly as biomarkers and as carriers of therapeutic molecules. With the use of genetic engineering and biomimetics, the field of exosome research for heart treatment is expected to solve various theranostic requirements in the field paving its way to the clinics.
Collapse
Affiliation(s)
- Sneha Yedavilli
- Department of Life Science, Central University of Karnataka, Kalaburagi, India
| | | | - Damini Singh
- Environmental Pollution Analysis Lab, Bhiwadi, India
| | - Rasmita Samal
- Department of Life Science, Central University of Karnataka, Kalaburagi, India
- *Correspondence: Rasmita Samal,
| |
Collapse
|
37
|
Matta A, Nader V, Lebrin M, Gross F, Prats AC, Cussac D, Galinier M, Roncalli J. Pre-Conditioning Methods and Novel Approaches with Mesenchymal Stem Cells Therapy in Cardiovascular Disease. Cells 2022; 11:1620. [PMID: 35626657 PMCID: PMC9140025 DOI: 10.3390/cells11101620] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) in the setting of cardiovascular disease, such as heart failure, cardiomyopathy and ischemic heart disease, has been associated with good clinical outcomes in several trials. A reduction in left ventricular remodeling, myocardial fibrosis and scar size, an improvement in endothelial dysfunction and prolonged cardiomyocytes survival were reported. The regenerative capacity, in addition to the pro-angiogenic, anti-apoptotic and anti-inflammatory effects represent the main target properties of these cells. Herein, we review the different preconditioning methods of MSCs (hypoxia, chemical and pharmacological agents) and the novel approaches (genetically modified MSCs, MSC-derived exosomes and engineered cardiac patches) suggested to optimize the efficacy of MSC therapy.
Collapse
Affiliation(s)
- Anthony Matta
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, 31059 Toulouse, France; (A.M.); (V.N.); (M.L.); (F.G.); (M.G.)
- Faculty of Medicine, Holy Spirit University of Kaslik, Kaslik 446, Lebanon
- Department of Cardiology, Intercommunal Hospital Centre Castres-Mazamet, 81100 Castres, France
| | - Vanessa Nader
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, 31059 Toulouse, France; (A.M.); (V.N.); (M.L.); (F.G.); (M.G.)
- Faculty of Pharmacy, Lebanese University, Beirut 6573/14, Lebanon
| | - Marine Lebrin
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, 31059 Toulouse, France; (A.M.); (V.N.); (M.L.); (F.G.); (M.G.)
- CIC-Biotherapies, University Hospital of Toulouse, 31059 Toulouse, France
| | - Fabian Gross
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, 31059 Toulouse, France; (A.M.); (V.N.); (M.L.); (F.G.); (M.G.)
- CIC-Biotherapies, University Hospital of Toulouse, 31059 Toulouse, France
| | | | - Daniel Cussac
- INSERM I2MC—UMR1297, 31432 Toulouse, France; (A.-C.P.); (D.C.)
| | - Michel Galinier
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, 31059 Toulouse, France; (A.M.); (V.N.); (M.L.); (F.G.); (M.G.)
| | - Jerome Roncalli
- Department of Cardiology, Institute CARDIOMET, University Hospital of Toulouse, 31059 Toulouse, France; (A.M.); (V.N.); (M.L.); (F.G.); (M.G.)
- CIC-Biotherapies, University Hospital of Toulouse, 31059 Toulouse, France
- INSERM I2MC—UMR1297, 31432 Toulouse, France; (A.-C.P.); (D.C.)
| |
Collapse
|
38
|
Wei J, Wang Z, Hou Z, Meng Y. The Influence of Empathy and Consumer Forgiveness on the Service Recovery Effect of Online Shopping. Front Psychol 2022; 13:842207. [PMID: 35432063 PMCID: PMC9007167 DOI: 10.3389/fpsyg.2022.842207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/24/2022] [Indexed: 11/30/2022] Open
Abstract
The service failure of online shopping has always plagued online stores, but the current academic circles still need to explore the service recovery of online shopping from the perspective of empathy and consumer forgiveness. Based on the service failure cases of real online shopping, this article uses the method of situational experiment to carry out empirical research, discusses the impact mechanism of service recovery effect from the perspective of empathy and consumer forgiveness, and tests the moderating role of online store reputation. The results show that in the online shopping service recovery scenario, empathy has a positive impact on consumer forgiveness, consumer forgiveness has a positive impact on consumer repurchase intention, and consumer forgiveness plays a mediating effect between empathy and consumer repurchase intention. Online store reputation plays a moderating role in the relationship between consumer forgiveness and consumer repurchase intention. The research conclusion of this article will help to expand the application of empathy and consumer forgiveness in the research of service recovery, enrich the theory of online shopping service recovery, improve the effect of online shopping service recovery, and promote the healthy development of online shopping business models.
Collapse
Affiliation(s)
- Jiahua Wei
- School of Business, Guilin University of Technology, Guilin, China
| | | | | | - Yongheng Meng
- School of Business, Guilin University of Technology, Guilin, China
| |
Collapse
|
39
|
Wu R, Fan X, Wang Y, Shen M, Zheng Y, Zhao S, Yang L. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Liver Immunity and Therapy. Front Immunol 2022; 13:833878. [PMID: 35309311 PMCID: PMC8930843 DOI: 10.3389/fimmu.2022.833878] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/15/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs), as the most common cell source for stem cell therapy, play an important role in the modulation of innate and adaptive immune responses and have been widely used in clinical trials to treat autoimmune and inflammatory diseases. Recent experimental and clinical studies have shown that MSC-derived extracellular vesicles (MSC-EVs) can inhibit the activation and proliferation of a variety of proinflammatory cells, such as Th1, Th17 and M1 macrophages, reducing the secretion of proinflammatory cytokines, while promoting the proliferation of anti-inflammatory cells, such as M2 macrophages and Tregs, and increasing the secretion of anti-inflammatory cytokines, thus playing a role in immune regulation and exhibiting immunomodulatory functions. Besides MSC-EVs are more convenient and less immunogenic than MSCs. There is growing interest in the role of MSC-EVs in liver diseases owing to the intrinsic liver tropism of MSC-EVs. In this review, we focus on the immunomodulatory effects of MSC-EVs and summarize the pivotal roles of MSC-EVs as a cell-free therapy in liver diseases, including NAFLD, AIH, acute liver failure, liver fibrosis and hepatic ischemia–reperfusion injury. Moreover, we provide a concise overview of the potential use and limits of MSC-EVs in clinical application.
Collapse
|
40
|
Song H, Chen S, Zhang T, Huang X, Zhang Q, Li C, Chen C, Chen S, Liu D, Wang J, Tu Y, Wu Y, Liu Y. Integrated Strategies of Diverse Feature Selection Methods Identify Aging-Based Reliable Gene Signatures for Ischemic Cardiomyopathy. Front Mol Biosci 2022; 9:805235. [PMID: 35300115 PMCID: PMC8921505 DOI: 10.3389/fmolb.2022.805235] [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: 10/30/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Objective: Ischemic cardiomyopathy (ICM) is a major cardiovascular state associated with prominently increased morbidity and mortality. Our purpose was to detect reliable gene signatures for ICM through integrated feature selection strategies.Methods: Transcriptome profiles of ICM were curated from the GEO project. Classification models, including least absolute shrinkage and selection operator (LASSO), support vector machine (SVM), and random forest, were adopted for identifying candidate ICM-specific genes for ICM. Immune cell infiltrates were estimated using the CIBERSORT method. Expressions of candidate genes were verified in ICM and healthy myocardial tissues via Western blotting. JC-1 staining, flow cytometry, and TUNEL staining were presented in hypoxia/reoxygenation (H/R)-stimulated H9C2 cells with TRMT5 deficiency.Results: Following the integration of three feature selection methods, we identified seven candidate ICM-specific genes including ASPN, TRMT5, LUM, FCN3, CNN1, PCNT, and HOPX. ROC curves confirmed the excellent diagnostic efficacy of this combination of previous candidate genes in ICM. Most of them presented prominent interactions with immune cell infiltrates. Their deregulations were confirmed in ICM than healthy myocardial tissues. TRMT5 expressions were remarkedly upregulated in H/R-stimulated H9C2 cells. TRMT5 deficiency enhanced mitochondrial membrane potential and reduced apoptosis in H/R-exposed H9C2 cells.Conclusion: Collectively, our findings identified reliable gene signatures through combination strategies of diverse feature selection methods, which facilitated the early detection of ICM and revealed the underlying mechanisms.
Collapse
Affiliation(s)
- Huafeng Song
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China
| | - Tingting Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofei Huang
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiyu Zhang
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Cuizhi Li
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunlin Chen
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaoxian Chen
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, School of Medicine, Guangdong Provincial People’s Hospital and Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, China
| | - Dehui Liu
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| | - Yingfeng Tu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| | - Yueheng Wu
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Cardiovascular Institute, School of Medicine, Guangdong Provincial People’s Hospital and Guangdong Academy of Medical Sciences, South China University of Technology, Guangzhou, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| | - Youbin Liu
- Department of Cardiology, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Jiawen Wang, ; Yingfeng Tu, ; Yueheng Wu, ; Youbin Liu,
| |
Collapse
|
41
|
Neef K, Drey F, Lepperhof V, Wahlers T, Hescheler J, Choi YH, Šarić T. Co-transplantation of Mesenchymal Stromal Cells and Induced Pluripotent Stem Cell-Derived Cardiomyocytes Improves Cardiac Function After Myocardial Damage. Front Cardiovasc Med 2022; 8:794690. [PMID: 35071360 PMCID: PMC8770928 DOI: 10.3389/fcvm.2021.794690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/22/2021] [Indexed: 01/01/2023] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) represent an attractive resource for cardiac regeneration. However, survival and functional integration of transplanted iPS-CM is poor and remains a major challenge for the development of effective therapies. We hypothesized that paracrine effects of co-transplanted mesenchymal stromal cells (MSCs) augment the retention and therapeutic efficacy of iPS-CM in a mouse model of myocardial infarction (MI). To test this, either iPS-CM, MSC, or both cell types were transplanted into the cryoinfarction border zone of syngeneic mice immediately after injury. Bioluminescence imaging (BLI) of iPS-CM did not confirm enhanced retention by co-application of MSC during the 28-day follow-up period. However, histological analyses of hearts 28 days after cell transplantation showed that MSC increased the fraction of animals with detectable iPS-CM by 2-fold. Cardiac MRI analyses showed that from day 14 after transplantation on, the animals that have received cells had a significantly higher left ventricular ejection fraction (LVEF) compared to the placebo group. There was no statistically significant difference in LVEF between animals transplanted only with iPS-CM or only with MSC. However, combined iPS-CM and MSC transplantation resulted in higher LVEF compared to transplantation of single-cell populations during the whole observation period. Histological analyses revealed that MSC increased the capillarization in the myocardium when transplanted alone or with iPS-CM and decreased the infarct scar area only when transplanted in combination with iPS-CM. These results indicate that co-transplantation of iPS-CM and MSC improves cardiac regeneration after cardiac damage, demonstrating the potential of combining multiple cell types for increasing the efficacy of future cardiac cell therapies.
Collapse
Affiliation(s)
- Klaus Neef
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Florian Drey
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Vera Lepperhof
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Clinic for Cardiac Surgery and Surgical Intensive Care Medicine, Kerckhoff Clinic Bad Nauheim, Kerckhoff Campus, Justus Liebig University Giessen, Giessen, Germany
| | - Tomo Šarić
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- *Correspondence: Tomo Šarić
| |
Collapse
|
42
|
HIF-1α overexpression in mesenchymal stem cell-derived exosome-encapsulated arginine-glycine-aspartate (RGD) hydrogels boost therapeutic efficacy of cardiac repair after myocardial infarction. Mater Today Bio 2021; 12:100171. [PMID: 34901821 PMCID: PMC8640519 DOI: 10.1016/j.mtbio.2021.100171] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 11/21/2022] Open
Abstract
Aims Naturally secreted extracellular vesicles (EVs) play important roles in stem-mediated cardioprotection. This study aimed to investigate the cardioprotective function and underlying mechanisms of EVs derived from HIF-1α engineered mesenchymal stem cells (MSCs) in a rat model of AMI. Methods and results EVs isolated from HIF-1α engineered MSCs (HIF-1α-EVs) and control MSCs (NC-EVs) were prepared. In in vitro experiments, the EVs were incubated with cardiomyocytes and endothelial cells exposed to hypoxia and serum deprivation (H/SD); in in vivo experiments, the EVs were injected in the acutely infarcted hearts of Sprague-Dawley rats. Compared with NC-EVs, HIF-1α-EVs significantly inhibited the apoptosis of cardiomyocytes and enhanced angiogenesis of endothelial cells; meanwhile, HIF-1α-EVs also significantly shrunk fibrotic area and strengthened cardiac function in infarcted rats. After treatment with EVs/RGD-biotin hydrogels, we observed longer retention, higher stability in HIF-1α-EVs, and stronger cardiac function in the rats. Quantitative real-time PCR (qRT-PCR) displayed that miRNA-221-3p was highly expressed in HIF-1α-EVs. After miR-221-3p was inhibited in HIF-1α-EVs, the biological effects of HIF-1α EVs on apoptosis and angiogenesis were attenuated. Conclusion EVs released by MSCs with HIF-1α overexpression can promote the angiogenesis of endothelial cells and the apoptosis of cardiomyocytes via upregulating the expression of miR-221-3p. RGD hydrogels can enhance the therapeutic efficacy of HIF-1α engineered MSCs-derived EVs.
Collapse
|
43
|
Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021; 11:187. [PMID: 34727974 PMCID: PMC8561357 DOI: 10.1186/s13578-021-00698-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapeutics is already available for treatment of a range of diseases or medical conditions. Autologous or allogeneic MSCs obtained from self or donors have their own advantages and disadvantages in their medical practice. Therapeutic benefits of using autologous vs. allogeneic MSCs are inconclusive. Transplanted MSCs within the body interact with their physical microenvironment or niche, physiologically or pathologically, and such cells in a newly established tissue microenvironment may be impacted by the pathological harmful environmental factors to alter their unique biological behaviors. Meanwhile, a temporary microenvironment/niche may be also altered by the resident or niche-surrounding MSCs. Therefore, the functional plasticity and heterogeneity of MSCs caused by different donors and subpopulations of MSCs may result in potential uncertainty in their safe and efficacious medical practice. Acknowledging a connection between MSCs' biology and their existing microenvironment, donor-controlled clinical practice for the long-term therapeutic benefit is suggested to further consider minimizing MSCs potential harm for MSC-based individual therapies. In this review, we summarize the advantages and disadvantages of autologous vs. allogeneic MSCs in their therapeutic applications. Among other issues, we highlight the importance of better understanding of the various microenvironments that may affect the properties of niche-surrounding MSCs and discuss the clinical applications of MSCs within different contexts for treatment of different diseases including cardiomyopathy, lupus and lupus nephritis, diabetes and diabetic complications, bone and cartilage repair, cancer and tissue fibrosis.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| | - Hua Zhao
- Institute of Reproductive Medicine, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Linna Cheng
- Institute of Hematology, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
44
|
Yamada S, Bartunek J, Behfar A, Terzic A. Mass Customized Outlook for Regenerative Heart Failure Care. Int J Mol Sci 2021; 22:11394. [PMID: 34768825 PMCID: PMC8583673 DOI: 10.3390/ijms222111394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/01/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Heart failure pathobiology is permissive to reparative intent. Regenerative therapies exemplify an emerging disruptive innovation aimed at achieving structural and functional organ restitution. However, mixed outcomes, complexity in use, and unsustainable cost have curtailed broader adoption, mandating the development of novel cardio-regenerative approaches. Lineage guidance offers a standardized path to customize stem cell fitness for therapy. A case in point is the molecular induction of the cardiopoiesis program in adult stem cells to yield cardiopoietic cell derivatives designed for heart failure treatment. Tested in early and advanced clinical trials in patients with ischemic heart failure, clinical grade cardiopoietic cells were safe and revealed therapeutic improvement within a window of treatment intensity and pre-treatment disease severity. With the prospect of mass customization, cardiopoietic guidance has been streamlined from the demanding, recombinant protein cocktail-based to a protein-free, messenger RNA-based single gene protocol to engineer affordable cardiac repair competent cells. Clinical trial biobanked stem cells enabled a systems biology deconvolution of the cardiopoietic cell secretome linked to therapeutic benefit, exposing a paracrine mode of action. Collectively, this new knowledge informs next generation regenerative therapeutics manufactured as engineered cellular or secretome mimicking cell-free platforms. Launching biotherapeutics tailored for optimal outcome and offered at mass production cost would contribute to advancing equitable regenerative care that addresses population health needs.
Collapse
Affiliation(s)
- Satsuki Yamada
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (S.Y.); (A.B.)
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jozef Bartunek
- Cardiovascular Center, OLV Hospital, 9300 Aalst, Belgium
| | - Atta Behfar
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (S.Y.); (A.B.)
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (S.Y.); (A.B.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
45
|
López-Díaz de Cerio A, Perez-Estenaga I, Inoges S, Abizanda G, Gavira JJ, Larequi E, Andreu E, Rodriguez S, Gil AG, Crisostomo V, Sanchez-Margallo FM, Bermejo J, Jauregui B, Quintana L, Fernández-Avilés F, Pelacho B, Prósper F. Preclinical Evaluation of the Safety and Immunological Action of Allogeneic ADSC-Collagen Scaffolds in the Treatment of Chronic Ischemic Cardiomyopathy. Pharmaceutics 2021; 13:pharmaceutics13081269. [PMID: 34452230 PMCID: PMC8399291 DOI: 10.3390/pharmaceutics13081269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/31/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022] Open
Abstract
The use of allogeneic adipose-derived mesenchymal stromal cells (alloADSCs) represents an attractive approach for treating myocardial infarction (MI). Furthermore, adding a natural support improves alloADSCs engraftment and survival in heart tissues, leading to a greater therapeutic effect. We aimed to examine the safety and immunological reaction induced by epicardial implantation of a clinical-grade collagen scaffold (CS) seeded with alloADSCs for its future application in humans. Thus, cellularized scaffolds were myocardially or subcutaneously implanted in immunosuppressed rodent models. The toxicological parameters were not significantly altered, and tumor formation was not found over the short or long term. Furthermore, biodistribution analyses in the infarcted immunocompetent rats displayed cell engraftment in the myocardium but no migration to other organs. The immunogenicity of alloADSC-CS was also evaluated in a preclinical porcine model of chronic MI; no significant humoral or cellular alloreactive responses were found. Moreover, CS cellularized with human ADSCs cocultured with human allogeneic immune cells produced no alloreactive response. Interestingly, alloADSC-CS significantly inhibited lymphocyte responses, confirming its immunomodulatory action. Thus, alloADSC-CS is likely safe and does not elicit any alloreactive immunological response in the host. Moreover, it exerts an immunomodulatory action, which supports its translation to a clinical setting.
Collapse
Affiliation(s)
- Ascensión López-Díaz de Cerio
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
| | - Iñigo Perez-Estenaga
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Susana Inoges
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
| | - Gloria Abizanda
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Juan José Gavira
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Department of Cardiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Eduardo Larequi
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Enrique Andreu
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
| | - Saray Rodriguez
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Ana Gloria Gil
- Department of Pharmacology and Toxicology, University of Navarra, 31009 Pamplona, Spain;
| | - Verónica Crisostomo
- Jesús Usón Minimally Invasive Surgery Centre (CCMIJU), Ctra. N-521, Km. 41.8, 10071 Cáceres, Spain; (V.C.); (F.M.S.-M.)
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
| | - Francisco Miguel Sanchez-Margallo
- Jesús Usón Minimally Invasive Surgery Centre (CCMIJU), Ctra. N-521, Km. 41.8, 10071 Cáceres, Spain; (V.C.); (F.M.S.-M.)
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
| | - Javier Bermejo
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | | | - Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Beatriz Pelacho
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
- Correspondence: (B.P.); (F.P.); Tel.: +34-948194700 (B.P.); +34-948255400 (F.P.)
| | - Felipe Prósper
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
- Correspondence: (B.P.); (F.P.); Tel.: +34-948194700 (B.P.); +34-948255400 (F.P.)
| |
Collapse
|
46
|
Comparison between Intra-Articular Injection of Infrapatellar Fat Pad (IPFP) Cell Concentrates and IPFP-Mesenchymal Stem Cells (MSCs) for Cartilage Defect Repair of the Knee Joint in Rabbits. Stem Cells Int 2021; 2021:9966966. [PMID: 34367294 PMCID: PMC8337123 DOI: 10.1155/2021/9966966] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic method in regenerative medicine. Our previous research adopted a simple nonenzymatic strategy for the preparation of a new type of ready-to-use infrapatellar fat pad (IPFP) cell concentrates. The aim of this study was to compare the therapeutic efficacy of intra-articular (IA) injection of autologous IPFP cell concentrates and allogeneic IPFP-MSCs obtained from these concentrates in a rabbit articular cartilage defect model. IPFP-MSCs sprouting from the IPFP cell concentrates were characterized via flow cytometry as well as based on their potential for differentiation into adipocytes, osteoblasts, and chondrocytes. In the rabbit model, cartilage defects were created on the trochlear groove, followed by treatment with IPFP cell concentrates, IPFP-MSCs, or normal saline IA injection. Distal femur samples were evaluated at 6 and 12 weeks posttreatment via macroscopic observation and histological assessment based on the International Cartilage Repair Society (ICRS) macroscopic scoring system as well as the ICRS visual histological assessment scale. The macroscopic score and histological score were significantly higher in the IPFP-MSC group compared to the IPFP cell concentrate group at 12 weeks. Further, both treatment groups had higher scores compared to the normal saline group. In comparison to the latter, the groups treated with IPFP-MSCs and IPFP cell concentrates showed considerably better cartilage regeneration. Overall, IPFP-MSCs represent an effective therapeutic strategy for stimulating articular cartilage regeneration. Further, due to the simple, cost-effective, nonenzymatic, and safe preparation process, IPFP cell concentrates may represent an effective alternative to stem cell-based therapy in the clinic.
Collapse
|