1
|
Huang Y, Hao X, Lin Z, Li L, Jiang H, Zhang H, Geng X, Zhu H, Wen H. Bio-distribution and toxicity potential of human umbilical cord mesenchymal stem cells in cynomolgus monkeys. Sci Rep 2024; 14:12251. [PMID: 38806615 PMCID: PMC11133417 DOI: 10.1038/s41598-024-63118-4] [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: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated promising advantages in the therapies of many diseases, while its multi-directional differentiation potential and immunotoxicity are the major concerns hindered their clinical translation. In this study, human umbilical Mesenchymal stem cell (hUC-MSCs) were labeled with a near-infrared fluorescent dye DiR before infused into cynomolgus monkeys, and the amount of hUC-MSCs in the peripheral blood were dynamically estimated from 5 min to 28 days post a single administration at 3 × 106 cells/kg and 2 × 107 cells/kg intravenously. As results, some hUC-MSCs distributed to the whole body within 5 min, while most of the cells accumulate in the lungs along with the systemic blood circulation, and subsequently released into the blood. The toxicity potentials of hUC-MSCs were investigated in another 30 cynomolgus monkeys, and the cells were repeatedly administrated at doses of 3 × 106 cells/kg and 2 × 107 cells/kg for 5 times on a weekly basis, with a recovery period of 1 months. hUC-MSCs showed no obvious toxic effects in cynomolgus monkeys, except xenogeneic immune rejection to human stem cells. Low levels of the hUC-MSC gene were detected in the peripheral blood of a few animals administered 2 × 107 cells/kg at 30 min subsequent to the first and last administration, and there was no significant difference in the copy number of the hUC-MSC gene in the blood samples compared with the first and last administration, indicating that the hUC-MSC was not significantly amplified in vivo, and it its safe in non-human primates. Our study for the first time verified the safety of long-term use of hUC-MSCs in primates. We have pioneered a technology for the real-time detection of hUC-MSCs in peripheral blood and provide dynamicand rapid monitoring of the distribution characteristics of hUC-MSCs in vivo. Here, we provide data supporting the application of such products for clinical treatment and the application of stem cells in major refractory diseases and regenerative medicine.
Collapse
Affiliation(s)
- Ying Huang
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Xiaofang Hao
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Zhi Lin
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Lulu Li
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Hua Jiang
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Hezhan Zhang
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Xingchao Geng
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China
| | - Hao Zhu
- Sinoneural Cell Engineering Group Co., Ltd., Shanghai, People's Republic of China.
| | - Hairuo Wen
- Key Laboratory of Beijing for Nonclinical Safety Evaluation Research of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, A8 Hongda Middle Road, Beijing, Economic-Technological Development Area, Beijing, 100176, People's Republic of China.
| |
Collapse
|
2
|
Whole-Heart Tissue Engineering and Cardiac Patches: Challenges and Promises. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010106. [PMID: 36671678 PMCID: PMC9855348 DOI: 10.3390/bioengineering10010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Despite all the advances in preventing, diagnosing, and treating cardiovascular disorders, they still account for a significant part of mortality and morbidity worldwide. The advent of tissue engineering and regenerative medicine has provided novel therapeutic approaches for the treatment of various diseases. Tissue engineering relies on three pillars: scaffolds, stem cells, and growth factors. Gene and cell therapy methods have been introduced as primary approaches to cardiac tissue engineering. Although the application of gene and cell therapy has resulted in improved regeneration of damaged cardiac tissue, further studies are needed to resolve their limitations, enhance their effectiveness, and translate them into the clinical setting. Scaffolds from synthetic, natural, or decellularized sources have provided desirable characteristics for the repair of cardiac tissue. Decellularized scaffolds are widely studied in heart regeneration, either as cell-free constructs or cell-seeded platforms. The application of human- or animal-derived decellularized heart patches has promoted the regeneration of heart tissue through in vivo and in vitro studies. Due to the complexity of cardiac tissue engineering, there is still a long way to go before cardiac patches or decellularized whole-heart scaffolds can be routinely used in clinical practice. This paper aims to review the decellularized whole-heart scaffolds and cardiac patches utilized in the regeneration of damaged cardiac tissue. Moreover, various decellularization methods related to these scaffolds will be discussed.
Collapse
|
3
|
Arenal Á, Ríos-Muñoz GR, Carta-Bergaz A, Ruiz-Hernández PM, Pérez-David E, Crisóstomo V, Loughlin G, Sanz-Ruiz R, Fernández-Portales J, Acosta A, Báez-Díaz C, Blanco-Blázquez V, Ledesma-Carbayo MJ, Pareja M, Fernández-Santos ME, Sánchez-Margallo FM, Casado JG, Fernández-Avilés F. Effects of Cardiac Stem Cell on Postinfarction Arrhythmogenic Substrate. Int J Mol Sci 2022; 23:16211. [PMID: 36555857 PMCID: PMC9781106 DOI: 10.3390/ijms232416211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Clinical data suggest that cardiosphere-derived cells (CDCs) could modify post-infarction scar and ventricular remodeling and reduce the incidence of ventricular tachycardia (VT). This paper assesses the effect of CDCs on VT substrate in a pig model of postinfarction monomorphic VT. We studied the effect of CDCs on the electrophysiological properties and histological structure of dense scar and heterogeneous tissue (HT). Optical mapping and histological evaluation were performed 16 weeks after the induction of a myocardial infarction by transient occlusion of the left anterior descending (LAD) artery in 21 pigs. Four weeks after LAD occlusion, pigs were randomized to receive intracoronary plus trans-myocardial CDCs (IC+TM group, n: 10) or to a control group. Optical mapping (OM) showed an action potential duration (APD) gradient between HT and normal tissue in both groups. CDCs increased conduction velocity (53 ± 5 vs. 45 ± 6 cm/s, p < 0.01), prolonged APD (280 ± 30 ms vs. 220 ± 40 ms, p < 0.01) and decreased APD dispersion in the HT. During OM, a VT was induced in one and seven of the IC+TM and control hearts (p = 0.03), respectively; five of these VTs had their critical isthmus located in intra-scar HT found adjacent to the coronary arteries. Histological evaluation of HT revealed less fibrosis (p < 0.01), lower density of myofibroblasts (p = 0.001), and higher density of connexin-43 in the IC+TM group. Scar and left ventricular volumes did not show differences between groups. Allogeneic CDCs early after myocardial infarction can modify the structure and electrophysiology of post-infarction scar. These findings pave the way for novel therapeutic properties of CDCs.
Collapse
Affiliation(s)
- Ángel Arenal
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Gonzalo R Ríos-Muñoz
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- BSEL-Biomedical Sciences and Engineering Laboratory, Bioengineering Department, Universidad Carlos III de Madrid, 28911 Madrid, Spain
| | - Alejandro Carta-Bergaz
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
| | - Pablo M Ruiz-Hernández
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Esther Pérez-David
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
| | - Verónica Crisóstomo
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain
| | - Gerard Loughlin
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | - Alejandra Acosta
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Claudia Báez-Díaz
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain
| | - Virginia Blanco-Blázquez
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain
| | - María J Ledesma-Carbayo
- Departamento Ingeniería Electrónica, Universidad Politécnica de Madrid and CIBER-BBN, 28040 Madrid, Spain
| | - Miriam Pareja
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
| | - María E Fernández-Santos
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
| | - Francisco M Sánchez-Margallo
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain
| | - Javier G Casado
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain
- Immunology Unit, University of Extremadura, 10003 Cáceres, Spain
| | - Francisco Fernández-Avilés
- Department of Cardiology, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Disease Network (CIBERCV), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
4
|
Báez-Díaz C, Blanco-Blázquez V, Sánchez-Margallo FM, López E, Martín H, Espona-Noguera A, Casado JG, Ciriza J, Pedraz JL, Crisóstomo V. Intrapericardial Delivery of APA-Microcapsules as Promising Stem Cell Therapy Carriers in an Experimental Acute Myocardial Infarction Model. Pharmaceutics 2021; 13:1824. [PMID: 34834235 PMCID: PMC8626005 DOI: 10.3390/pharmaceutics13111824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/15/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023] Open
Abstract
The administration of cardiosphere-derived cells (CDCs) after acute myocardial infarction (AMI) is very promising. CDC encapsulation in alginate-poly-l-lysine-alginate (APA) could increase cell survival and adherence. The intrapericardial (IP) approach potentially achieves high concentrations of the therapeutic agent in the infarcted area. We aimed to evaluate IP therapy using a saline vehicle as a control (CON), a dose of 30 × 106 CDCs (CDCs) or APA microcapsules containing 30 × 106 CDCs (APA-CDCs) at 72 h in a porcine AMI model. Magnetic resonance imaging (MRI) was used to determine the left ventricular ejection fraction (LVEF), infarct size (IS), and indexed end diastolic and systolic volumes (EDVi; ESVi) pre- and 10 weeks post-injection. Programmed electrical stimulation (PES) was performed to test arrhythmia inducibility before euthanasia. Histopathological analysis was carried out afterwards. The IP infusion was successful in all animals. At 10 weeks, MRI revealed significantly higher LVEF in the APA-CDC group compared with CON. No significant differences were observed among groups in IS, EDVi, ESVi, PES and histopathological analyses. In conclusion, the IP injection of CDCs (microencapsulated or not) was feasible and safe 72 h post-AMI in the porcine model. Moreover, CDCs APA encapsulation could have a beneficial effect on cardiac function, reflected by a higher LVEF at 10 weeks.
Collapse
Affiliation(s)
- Claudia Báez-Díaz
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Virginia Blanco-Blázquez
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Francisco Miguel Sánchez-Margallo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Esther López
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Helena Martín
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| | - Albert Espona-Noguera
- Centro de Investigaciones y Estudios Avanzados Lucio Lascaray (CIEA), Laboratorio de Desarrollo y Evaluación de Medicamentos, 01006 Vitoria Gasteiz, Spain; (A.E.-N.); (J.L.P.)
- CIBER bbn, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Javier G. Casado
- Immunology Unit-Institute of Molecular Pathology Biomarkers, Veterinary Faculty, University of Extremadura, 10003 Cáceres, Spain;
| | - Jesús Ciriza
- CIBER bbn, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
| | - José Luis Pedraz
- Centro de Investigaciones y Estudios Avanzados Lucio Lascaray (CIEA), Laboratorio de Desarrollo y Evaluación de Medicamentos, 01006 Vitoria Gasteiz, Spain; (A.E.-N.); (J.L.P.)
- CIBER bbn, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Verónica Crisóstomo
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (V.B.-B.); (F.M.S.-M.); (V.C.)
- Fundación Centro de Cirugía de Mínima Invasión Jesús Usón, 10071 Cáceres, Spain; (E.L.); (H.M.)
| |
Collapse
|
5
|
Chirurgische Beiträge zur kardialen Stammzelltherapie. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2021. [DOI: 10.1007/s00398-021-00454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
6
|
Zeng CY, Xu J, Liu X, Lu YQ. Cardioprotective Roles of Endothelial Progenitor Cell-Derived Exosomes. Front Cardiovasc Med 2021; 8:717536. [PMID: 34513956 PMCID: PMC8428070 DOI: 10.3389/fcvm.2021.717536] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
With the globally increasing prevalence, cardiovascular diseases (CVDs) have become the leading cause of mortality. The transplantation of endothelial progenitor cells (EPCs) holds a great promise due to their potential for vasculogenesis, angiogenesis, and protective cytokine release, whose mechanisms are essential for CVD therapies. In reality, many investigations have attributed the therapeutic effects of EPC transplantation to the secretion of paracrine factors rather than the differentiation function. Of note, previous studies have suggested that EPCs could also release exosomes (diameter range of 30–150 nm), which carry various lipids and proteins and are abundant in microRNAs. The EPC-derived exosomes (EPC-EXs) were reported to act on the heart and blood vessels and were implicated in anti-inflammation, anti-oxidation, anti-apoptosis, the inhibition of endothelial-to-mesenchymal transition (EndMT), and cardiac fibrosis, as well as anti-vascular remodeling and angiogenesis, which were considered as protective effects against CVDs. In this review, we summarize the current knowledge on using EPC-EXs as therapeutic agents and provide a detailed description of their identified mechanisms of action to promote the prognosis of CVDs.
Collapse
Affiliation(s)
- Cai-Yu Zeng
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jia Xu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xin Liu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuan-Qiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Geriatrics, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
7
|
Yao J, Huang K, Zhu D, Chen T, Jiang Y, Zhang J, Mi L, Xuan H, Hu S, Li J, Zhou Y, Cheng K. A Minimally Invasive Exosome Spray Repairs Heart after Myocardial Infarction. ACS NANO 2021; 15:11099-11111. [PMID: 34152126 DOI: 10.1021/acsnano.1c00628] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Myocardial infarction (MI) remains the most common cause of death worldwide. Many MI survivors will suffer from recurrent heart failure (HF), which has been recognized as a determinant of adverse prognosis. Despite the success of improved early survival after MI by primary percutaneous coronary intervention, HF after MI is becoming the major driver of late morbidity, mortality, and healthcare costs. The development of regenerative medicine has brought hope to MI treatment in the past decade. Mesenchymal stem cell (MSC)-derived exosomes have been established as an essential part of stem cell paracrine factors for heart regeneration. However, its regenerative power is hampered by low delivery efficiency to the heart. We designed, fabricated, and tested a minimally invasive exosome spray (EXOS) based on MSC exosomes and biomaterials. In a mouse model of acute myocardial infarction, EXOS improved cardiac function and reduced fibrosis, and promoted endogenous angiomyogenesis in the post-injury heart. We further tested the feasibility and safety of EXOS in a pig model. Our results indicate that EXOS is a promising strategy to deliver therapeutic exosomes for heart repair.
Collapse
Affiliation(s)
- Jialu Yao
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Tan Chen
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Junyi Zhang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Lijie Mi
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - He Xuan
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Junlang Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Yafeng Zhou
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000, China
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
8
|
Das Ghosh L, Hasan J, Jain A, Sundaresan NR, Chatterjee K. A nanopillar array on black titanium prepared by reactive ion etching augments cardiomyogenic commitment of stem cells. NANOSCALE 2019; 11:20766-20776. [PMID: 31651003 DOI: 10.1039/c9nr03424b] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A major impediment in the clinical translation of stem cell therapy has been the inability to efficiently and reproducibly direct differentiation of a large population of stem cells. Thus, we aimed to engineer a substrate for culturing stem cells to efficiently induce cardiomyogenic lineage commitment. In this work, we present a nanopillar array on the surface of titanium that was prepared by mask-less reactive ion etching. Scanning electron and atomic force microscopy revealed that the surface was covered by vertically aligned nanopillars each of ≈1 μm with a diameter of ≈80 nm. The nanopillars supported the attachment and proliferation of human mesenchymal stem cells (hMSCs). Cardiomyogenic lineage commitment of the stem cells was more enhanced on the nanopillars than on the smooth surface. When co-cultured with neonatal rat cardiomyocytes, the cyclic pattern of calcium transport observed distinctly in cells differentiated on the arrays compared to the cells cultured on the smooth surface was the functional validation of differentiation. The use of small molecule inhibitors revealed that integrins namely, α2β1 and αvβ3, are essential for cardiomyogenesis on the nanostructured surface, which is further mediated by FAK, Erk and Akt cell signaling pathways. This study demonstrates that the nanopillar array efficiently promotes the cardiomyogenic lineage commitment of stem cells via integrin-mediated signaling and can potentially serve as a platform for the ex vivo differentiation of stem cells toward cell therapy in cardiac tissue repair and regeneration.
Collapse
Affiliation(s)
- Lopamudra Das Ghosh
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Jafar Hasan
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Aditi Jain
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| | - Nagalingam R Sundaresan
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India. and Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India. and Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
9
|
Li X, Xiang D, Shu Y, Zeng X, Li Y. Mitigating effect of tanshinone IIA on ventricular remodeling in rats with pressure overload-induced heart failure. Acta Cir Bras 2019; 34:e201900807. [PMID: 31618407 PMCID: PMC6802940 DOI: 10.1590/s0102-865020190080000007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 07/11/2019] [Indexed: 11/22/2022] Open
Abstract
Purpose To investigate the effect of tanshinone IIA (TIIA) on ventricular remodeling in rats with pressure overload-induced heart failure. Methods Pressure overload-induced heart failure model (abdominal aortic coarctation) was established in 40 rats, which were divided into model and 5, 10 and 20 mg/kg TIIA groups. Ten rats receiving laparotomy excepting abdominal aortic coarctation were enrolled in sham-operated group. The 5, 10 and 20 mg/kg TIIA groups were treated with 5, 10 and 20 mg/kg TIIA, respectively, for 8 weeks. Results Compared with model group, in 20 mg/kg TIIA group the left ventricular ejection fraction, left ventricular fractional shortening, left ventricular systolic pressure, ±maximum left ventricular pressure rising and dropping rate, and myocardial B-cell lymphoma-2 and cleaved cysteinyl aspartate specific proteinase-3 protein levels were increased, respectively (P<0.05), and the left ventricular end diastolic diameter, left ventricular end systolic diameter, left ventricular end diastolic pressure, heart weight index, left ventricular weight index, serum B-type brain natriuretic peptide, interleukin 6 and C-reactive protein levels and myocardial B-cell lymphoma-2 associated X protein level were decreased, respectively (P<0.05). Conclusion TIIA may alleviate ventricular remodeling in rats with pressure overload-induced heart failure heart by reducing inflammatory response and cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Xu Li
- Master, Department of Cardiac Surgery , Guizhou Provincial People's Hospital , Guiyang , China . Design of the study, final approval
| | - Daokang Xiang
- MD, Department of Cardiac Surgery , Guizhou Provincial People's Hospital , Guiyang , China . Design of the study, critical revision, final approval
| | - Yizhu Shu
- MD, Department of Cardiac Surgery , Guizhou Provincial People's Hospital , Guiyang , China . Conception of the study, final approval
| | - Xiangjun Zeng
- MD, Department of Cardiac Surgery , Guizhou Provincial People's Hospital , Guiyang , China . Acquisition of data, statistical analyses, final approval
| | - Yonghong Li
- MD, Department of Cardiac Surgery , Guizhou Provincial People's Hospital , Guiyang , China . Manuscript writing, final approval
| |
Collapse
|
10
|
Fernandes GC, Fernandes ADF, Rivera M, Khan A, Schulman IH, Lambrakos LK, Myerburg RJ, Goldberger JJ, Hare JM, Mitrani RD. A meta‐analysis of arrhythmia endpoints in randomized controlled trials of transendocardial stem cell injections for chronic ischemic heart disease. J Cardiovasc Electrophysiol 2019; 30:2492-2500. [DOI: 10.1111/jce.14185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/02/2019] [Accepted: 09/11/2019] [Indexed: 02/01/2023]
Affiliation(s)
- Gilson C. Fernandes
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | | | - Manuel Rivera
- Division of Cardiology and Cardiovascular Washington University in Saint Louis Saint Louis Missouri
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Ivonne H. Schulman
- Department of Medicine University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Litsa K. Lambrakos
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | - Robert J. Myerburg
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | | | - Joshua M. Hare
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Raul D. Mitrani
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
11
|
Poglajen G, Zemljič G, Cerar A, Frljak S, Jaklič M, Androcec V, Vrtovec B. Transendocardial CD34+ Cell Therapy does not Increase the Risk of Ventricular Arrhythmias in Patients with Chronic Heart Failure. Cell Transplant 2019; 28:856-863. [PMID: 31046425 PMCID: PMC6719496 DOI: 10.1177/0963689719840351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ventricular arrhythmias (VA) are of major concern in the field of cell therapy, potentially limiting its safety and efficacy. We sought to investigate the effects of CD34+ cell therapy on VA burden in patients with chronic heart failure (CHF). We performed registry data analysis of patients with CHF and implanted ICD/CRT devices treated with transendocardial CD 34+ cell therapy. Demographic, echocardiographic, and biochemical parameters were analyzed. Device records were reviewed and the number and type of VA 1 year prior to and 1 year after cell therapy were analyzed. All patients underwent electroanatomical mapping, and myocardial scar was defined as unipolar voltage (UV) <8.3 mV and linear local shortening (LLS) <6%. Of 209 patients screened, 48 met inclusion criteria. The mean age of the patients was 52 years and 88% were male. Nonischemic and ischemic cardiomyopathy were present in 55% and 45% of patients. The average serum creatinine was 91±26 µmol/L, serum bilirubin 18±9 µmol/L, NT-proBNP 1767 (468, 2446) pg/mL, LVEF 27±9% and 6’ walk test 442±123 m. The average scar burden in patients with nonischemic and ischemic DCM was 58±15% and 51±25% (P=0.48). No significant difference in VA burden was observed before and after cell therapy (48% vs. 44%; P=0.68). ICD activation occurred in 19% and 27% of patients before and after cell therapy (P=0.33). According to our results, transendocardial CD34+ cell therapy does not appear to increase the risk of VA in chronic heart failure patients.
Collapse
Affiliation(s)
- Gregor Poglajen
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia.,2 Faculy of Medicine, Ljubljana, Slovenia
| | - Gregor Zemljič
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Andraž Cerar
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Sabina Frljak
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Martina Jaklič
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Vesna Androcec
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Bojan Vrtovec
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia.,2 Faculy of Medicine, Ljubljana, Slovenia
| |
Collapse
|
12
|
Liang J, Huang W, Jiang L, Paul C, Li X, Wang Y. Concise Review: Reduction of Adverse Cardiac Scarring Facilitates Pluripotent Stem Cell-Based Therapy for Myocardial Infarction. Stem Cells 2019; 37:844-854. [PMID: 30913336 PMCID: PMC6599570 DOI: 10.1002/stem.3009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
Pluripotent stem cells (PSCs) are an attractive, reliable source for generating functional cardiomyocytes for regeneration of infarcted heart. However, inefficient cell engraftment into host tissue remains a notable challenge to therapeutic success due to mechanical damage or relatively inhospitable microenvironment. Evidence has shown that excessively formed scar tissues around cell delivery sites present as mechanical and biological barriers that inhibit migration and engraftment of implanted cells. In this review, we focus on the functional responses of stem cells and cardiomyocytes during the process of cardiac fibrosis and scar formation. Survival, migration, contraction, and coupling function of implanted cells may be affected by matrix remodeling, inflammatory factors, altered tissue stiffness, and presence of electroactive myofibroblasts in the fibrotic microenvironment. Although paracrine factors from implanted cells can improve cardiac fibrosis, the transient effect is insufficient for complete repair of an infarcted heart. Furthermore, investigation of interactions between implanted cells and fibroblasts including myofibroblasts helps the identification of new targets to optimize the host substrate environment for facilitating cell engraftment and functional integration. Several antifibrotic approaches, including the use of pharmacological agents, gene therapies, microRNAs, and modified biomaterials, can prevent progression of heart failure and have been developed as adjunct therapies for stem cell-based regeneration. Investigation and optimization of new biomaterials is also required to enhance cell engraftment of engineered cardiac tissue and move PSCs from a laboratory setting into translational medicine.
Collapse
Affiliation(s)
- Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xiangnan Li
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
13
|
Onoshima D, Yukawa H, Baba Y. Nanobiodevices for Cancer Diagnostics and Stem Cell Therapeutics. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
14
|
Huang K, Hu S, Cheng K. A New Era of Cardiac Cell Therapy: Opportunities and Challenges. Adv Healthc Mater 2019; 8:e1801011. [PMID: 30548836 PMCID: PMC6368830 DOI: 10.1002/adhm.201801011] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Indexed: 12/15/2022]
Abstract
Myocardial infarction (MI), caused by coronary heart disease (CHD), remains one of the most common causes of death in the United States. Over the last few decades, scientists have invested considerable resources on the study and development of cell therapies for myocardial regeneration after MI. However, due to a number of limitations, they are not yet readily available for clinical applications. Mounting evidence supports the theory that paracrine products are the main contributors to the regenerative effects attributed to these cell therapies. The next generation of cell-based MI therapies will identify and isolate cell products and derivatives, integrate them with biocompatible materials and technologies, and use them for the regeneration of damaged myocardial tissue. This review discusses the progress made thus far in pursuit of this new generation of cell therapies. Their fundamental regenerative mechanisms, their potential to combine with other therapeutic products, and their role in shaping new clinical approaches for heart tissue engineering, are addressed.
Collapse
Affiliation(s)
- Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27607, USA
- Pharmacoengineeirng and Molecular Pharmaceutics Division, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
15
|
Cell-Based Therapies for Cardiac Regeneration: A Comprehensive Review of Past and Ongoing Strategies. Int J Mol Sci 2018; 19:ijms19103194. [PMID: 30332812 PMCID: PMC6214096 DOI: 10.3390/ijms19103194] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022] Open
Abstract
Despite considerable improvements in the treatment of cardiovascular diseases, heart failure (HF) still represents one of the leading causes of death worldwide. Poor prognosis is mostly due to the limited regenerative capacity of the adult human heart, which ultimately leads to left ventricular dysfunction. As a consequence, heart transplantation is virtually the only alternative for many patients. Therefore, novel regenerative approaches are extremely needed, and several attempts have been performed to improve HF patients’ clinical conditions by promoting the replacement of the lost cardiomyocytes and by activating cardiac repair. In particular, cell-based therapies have been shown to possess a great potential for cardiac regeneration. Different cell types have been extensively tested in clinical trials, demonstrating consistent safety results. However, heterogeneous efficacy data have been reported, probably because precise end-points still need to be clearly defined. Moreover, the principal mechanism responsible for these beneficial effects seems to be the paracrine release of antiapoptotic and immunomodulatory molecules from the injected cells. This review covers past and state-of-the-art strategies in cell-based heart regeneration, highlighting the advantages, challenges, and limitations of each approach.
Collapse
|
16
|
Loisel F, Provost B, Haddad F, Guihaire J, Amsallem M, Vrtovec B, Fadel E, Uzan G, Mercier O. Stem cell therapy targeting the right ventricle in pulmonary arterial hypertension: is it a potential avenue of therapy? Pulm Circ 2018; 8:2045893218755979. [PMID: 29480154 PMCID: PMC5844533 DOI: 10.1177/2045893218755979] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an incurable disease characterized by an increase in pulmonary arterial pressure due to pathological changes to the pulmonary vascular bed. As a result, the right ventricle (RV) is subject to an increased afterload and undergoes multiple changes, including a decrease in capillary density. All of these dysfunctions lead to RV failure. A number of studies have shown that RV function is one of the main prognostic factors for PAH patients. Many stem cell therapies targeting the left ventricle are currently undergoing development. The promising results observed in animal models have led to clinical trials that have shown an improvement of cardiac function. In contrast to left heart disease, stem cell therapy applied to the RV has remained poorly studied, even though it too may provide a therapeutic benefit. In this review, we discuss stem cell therapy as a treatment for RV failure in PAH. We provide an overview of the results of preclinical and clinical studies for RV cell therapies. Although a large number of studies have targeted the pulmonary circulation rather than the RV directly, there are nonetheless encouraging results in the literature that indicate that cell therapies may have a direct beneficial effect on RV function. This cell therapy strategy may therefore hold great promise and warrants further studies in PAH patients.
Collapse
Affiliation(s)
- Fanny Loisel
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,2 Inserm 1197 Research Unit, Universite Paris Sud, Paris-Saclay University, Villejuif, France
| | - Bastien Provost
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - François Haddad
- 3 Cardiovascular Medicine, Stanford Hospital, Stanford University, CA, USA
| | - Julien Guihaire
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Myriam Amsallem
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Bojan Vrtovec
- 4 Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Elie Fadel
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,5 Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Georges Uzan
- 2 Inserm 1197 Research Unit, Universite Paris Sud, Paris-Saclay University, Villejuif, France
| | - Olaf Mercier
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,5 Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| |
Collapse
|
17
|
Melly L, Cerino G, Frobert A, Cook S, Giraud MN, Carrel T, Tevaearai Stahel HT, Eckstein F, Rondelet B, Marsano A, Banfi A. Myocardial infarction stabilization by cell-based expression of controlled Vascular Endothelial Growth Factor levels. J Cell Mol Med 2018; 22:2580-2591. [PMID: 29478261 PMCID: PMC5908097 DOI: 10.1111/jcmm.13511] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/23/2017] [Indexed: 01/24/2023] Open
Abstract
Vascular Endothelial Growth Factor (VEGF) can induce normal or aberrant angiogenesis depending on the amount secreted in the microenvironment around each cell. Towards a possible clinical translation, we developed a Fluorescence Activated Cell Sorting (FACS)-based technique to rapidly purify transduced progenitors that homogeneously express a desired specific VEGF level from heterogeneous primary populations. Here, we sought to induce safe and functional angiogenesis in ischaemic myocardium by cell-based expression of controlled VEGF levels. Human adipose stromal cells (ASC) were transduced with retroviral vectors and FACS purified to generate two populations producing similar total VEGF doses, but with different distributions: one with cells homogeneously producing a specific VEGF level (SPEC), and one with cells heterogeneously producing widespread VEGF levels (ALL), but with an average similar to that of the SPEC population. A total of 70 nude rats underwent myocardial infarction by coronary artery ligation and 2 weeks later VEGF-expressing or control cells, or saline were injected at the infarction border. Four weeks later, ventricular ejection fraction was significantly worsened with all treatments except for SPEC cells. Further, only SPEC cells significantly increased the density of homogeneously normal and mature microvascular networks. This was accompanied by a positive remodelling effect, with significantly reduced fibrosis in the infarcted area. We conclude that controlled homogeneous VEGF delivery by FACS-purified transduced ASC is a promising strategy to achieve safe and functional angiogenesis in myocardial ischaemia.
Collapse
Affiliation(s)
- Ludovic Melly
- Cell and Gene Therapy, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland.,Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland.,Department of Cardiac Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Giulia Cerino
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Aurélien Frobert
- Department of Cardiology, University of Fribourg, Fribourg, Switzerland
| | - Stéphane Cook
- Department of Cardiology, University of Fribourg, Fribourg, Switzerland
| | | | - Thierry Carrel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Hendrik T Tevaearai Stahel
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Friedrich Eckstein
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Benoît Rondelet
- Department of Cardiac Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Anna Marsano
- Cardiac Surgery and Engineering, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| | - Andrea Banfi
- Cell and Gene Therapy, Departments of Biomedicine and Surgery, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
18
|
Peña B, Bosi S, Aguado BA, Borin D, Farnsworth NL, Dobrinskikh E, Rowland TJ, Martinelli V, Jeong M, Taylor MRG, Long CS, Shandas R, Sbaizero O, Prato M, Anseth KS, Park D, Mestroni L. Injectable Carbon Nanotube-Functionalized Reverse Thermal Gel Promotes Cardiomyocytes Survival and Maturation. ACS APPLIED MATERIALS & INTERFACES 2017; 9:31645-31656. [PMID: 28895403 PMCID: PMC5672802 DOI: 10.1021/acsami.7b11438] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The ability of the adult heart to regenerate cardiomyocytes (CMs) lost after injury is limited, generating interest in developing efficient cell-based transplantation therapies. Rigid carbon nanotubes (CNTs) scaffolds have been used to improve CMs viability, proliferation, and maturation, but they require undesirable invasive surgeries for implantation. To overcome this limitation, we developed an injectable reverse thermal gel (RTG) functionalized with CNTs (RTG-CNT) that transitions from a solution at room temperature to a three-dimensional (3D) gel-based matrix shortly after reaching body temperature. Here we show experimental evidence that this 3D RTG-CNT system supports long-term CMs survival, promotes CMs alignment and proliferation, and improves CMs function when compared with traditional two-dimensional gelatin controls and 3D plain RTG system without CNTs. Therefore, our injectable RTG-CNT system could potentially be used as a minimally invasive tool for cardiac tissue engineering efforts.
Collapse
Affiliation(s)
- Brisa Peña
- Cardiovascular Institute, University of Colorado Denver Anschutz Medical Campus, School of Medicine, Division of Cardiology, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Susanna Bosi
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste 34127, Italy
| | - Brian A. Aguado
- Department of Chemical and Biological Engineering and Howard Hughes Medical Institute and the BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309, United States
| | - Daniele Borin
- Department of Engineering and Architecture, University of Trieste, Trieste 34127, Italy
| | - Nikki L. Farnsworth
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Evgenia Dobrinskikh
- Department of Medicine, University of Colorado Denver Anschutz Medical Campus, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Teisha J. Rowland
- Cardiovascular Institute, University of Colorado Denver Anschutz Medical Campus, School of Medicine, Division of Cardiology, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Valentina Martinelli
- International Center for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, Trieste 34149, Italy
| | - Mark Jeong
- Cardiovascular Institute, University of Colorado Denver Anschutz Medical Campus, School of Medicine, Division of Cardiology, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Matthew R. G. Taylor
- Cardiovascular Institute, University of Colorado Denver Anschutz Medical Campus, School of Medicine, Division of Cardiology, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Carlin S. Long
- Cardiovascular Institute, University of Colorado Denver Anschutz Medical Campus, School of Medicine, Division of Cardiology, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| | - Robin Shandas
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Trieste 34127, Italy
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste 34127, Italy
- Carbon Nanobiotechnology Laboratory, CIC biomaGUNE, Paseo de Miramón 182 20009, Donostia-San Sebastián 20009, Spain
- Basque Foundation for Science, Ikerbasque, Bilbao 48013, Spain
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering and Howard Hughes Medical Institute and the BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309, United States
| | - Daewon Park
- Bioengineering Department, University of Colorado Denver Anschutz Medical Campus, Bioscience 2 1270 E. Montview Avenue, Suite 100, Aurora, Colorado 80045, United States
| | - Luisa Mestroni
- Cardiovascular Institute, University of Colorado Denver Anschutz Medical Campus, School of Medicine, Division of Cardiology, 12700 E. 19th Avenue, Bldg. P15, Aurora, Colorado 80045, United States
| |
Collapse
|
19
|
Zhang L, Li X, Yu X, Li Y, Sun A, Huang C, Xu F, Guo J, Sun Y, Zhang X, Yang X, Zhang C. Construction of vascularized pacemaker tissues by seeding cardiac progenitor cells and endothelial progenitor cells into Matrigel. Life Sci 2017; 179:139-146. [DOI: 10.1016/j.lfs.2017.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 01/05/2023]
|
20
|
Petchdee S, Sompeewong S. Intravenous administration of puppy deciduous teeth stem cells in degenerative valve disease. Vet World 2016; 9:1429-1434. [PMID: 28096616 PMCID: PMC5234058 DOI: 10.14202/vetworld.2016.1429-1434] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 11/16/2016] [Indexed: 01/05/2023] Open
Abstract
Aim: The objective of this study is to investigate the improvement of heart function in dogs with chronic valvular heart disease after puppy deciduous teeth stem cells (pDSCs) administration. Materials and Methods: 20 client-owned dogs with degenerative valvular heart disease underwent multiple intravenous injections of allogeneic pDSCs. Dogs were randomly assigned to two groups: (i) Control group (n=10) with standard treatment for heart failure and (ii) group with standard treatment and multiple administrations of pDSCs (n=10). Electrocardiography, complete transthoracic echocardiography, thoracic radiography, and blood pressure were recorded before and after pDSCs injections for 15, 30 and 60 days. Results: Post pDSCs injection showed measurable improvement in left ventricular ejection fraction, American College of Veterinary Internal Medicine (ACVIM) functional class significantly improved and improved quality of life scores were observed. In the control group, there were no significant enhancements in heart function or ACVIM class. Conclusions: This finding suggests that pDSCs could be a supplement for valvular heart disease treatment.
Collapse
Affiliation(s)
- Soontaree Petchdee
- Department of Large Animal and Wildlife Clinical Science, Faculty of Veterinary Medicine, Kasetsart University, Kamphaengsaen, Nakhorn Pathom 73140, Thailand
| | - Sarunya Sompeewong
- Kasetsart University, Veterinary Teaching Animal Hospital, Kamphaeng Saen, Thailand
| |
Collapse
|
21
|
Abstract
OPINION STATEMENT Despite significant advances in the treatment of ischemic heart disease (IHD), it remains the leading cause of mortality worldwide. Undoubtedly, methods for regenerating the injured human heart are urgently needed, and whilst exciting progress has been made from utilizing stem cell therapy for cardiac regeneration, several major challenges still remain. In particular, one major safety issue is the occurrence of potentially life-threatening ventricular arrhythmias after cell therapy. Several drivers may be responsible for this, ranging from the potential inherent arrhythmogenicity of delivered stem cells to that of the underlying IHD. Therefore, it is imperative to thoroughly assess the risk-to-benefit ratio of such treatments prior to the clinical application. As such, despite the considerable progress made in stem cell therapy over the past decades, many obstacles still lie ahead.
Collapse
|
22
|
Richards DJ, Tan Y, Coyle R, Li Y, Xu R, Yeung N, Parker A, Menick DR, Tian B, Mei Y. Nanowires and Electrical Stimulation Synergistically Improve Functions of hiPSC Cardiac Spheroids. NANO LETTERS 2016; 16:4670-8. [PMID: 27328393 PMCID: PMC4994528 DOI: 10.1021/acs.nanolett.6b02093] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The advancement of human induced pluripotent stem-cell-derived cardiomyocyte (hiPSC-CM) technology has shown promising potential to provide a patient-specific, regenerative cell therapy strategy to treat cardiovascular disease. Despite the progress, the unspecific, underdeveloped phenotype of hiPSC-CMs has shown arrhythmogenic risk and limited functional improvements after transplantation. To address this, tissue engineering strategies have utilized both exogenous and endogenous stimuli to accelerate the development of hiPSC-CMs. Exogenous electrical stimulation provides a biomimetic pacemaker-like stimuli that has been shown to advance the electrical properties of tissue engineered cardiac constructs. Recently, we demonstrated that the incorporation of electrically conductive silicon nanowires to hiPSC cardiac spheroids led to advanced structural and functional development of hiPSC-CMs by improving the endogenous electrical microenvironment. Here, we reasoned that the enhanced endogenous electrical microenvironment of nanowired hiPSC cardiac spheroids would synergize with exogenous electrical stimulation to further advance the functional development of nanowired hiPSC cardiac spheroids. For the first time, we report that the combination of nanowires and electrical stimulation enhanced cell-cell junction formation, improved development of contractile machinery, and led to a significant decrease in the spontaneous beat rate of hiPSC cardiac spheroids. The advancements made here address critical challenges for the use of hiPSC-CMs in cardiac developmental and translational research and provide an advanced cell delivery vehicle for the next generation of cardiac repair.
Collapse
Affiliation(s)
- Dylan J. Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yang Li
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ruoyu Xu
- Department of Chemistry, the James Franck Institute and the Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | - Nelson Yeung
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Arran Parker
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Donald R. Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston SC 29425, USA
| | - Bozhi Tian
- Department of Chemistry, the James Franck Institute and the Institute for Biophysical Dynamics, the University of Chicago, Chicago, IL 60637, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
23
|
Garbayo E, Gavira JJ, de Yebenes MG, Pelacho B, Abizanda G, Lana H, Blanco-Prieto MJ, Prosper F. Catheter-based Intramyocardial Injection of FGF1 or NRG1-loaded MPs Improves Cardiac Function in a Preclinical Model of Ischemia-Reperfusion. Sci Rep 2016; 6:25932. [PMID: 27184924 PMCID: PMC4868965 DOI: 10.1038/srep25932] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/25/2016] [Indexed: 01/05/2023] Open
Abstract
Cardiovascular protein therapeutics such as neuregulin (NRG1) and acidic-fibroblast growth factor (FGF1) requires new formulation strategies that allow for sustained bioavailability of the drug in the infarcted myocardium. However, there is no FDA-approved injectable protein delivery platform due to translational concerns about biomaterial administration through cardiac catheters. We therefore sought to evaluate the efficacy of percutaneous intramyocardial injection of poly(lactic-co-glycolic acid) microparticles (MPs) loaded with NRG1 and FGF1 using the NOGA MYOSTAR injection catheter in a porcine model of ischemia-reperfusion. NRG1- and FGF1-loaded MPs were prepared using a multiple emulsion solvent-evaporation technique. Infarcted pigs were treated one week after ischemia-reperfusion with MPs containing NRG1, FGF1 or non-loaded MPs delivered via clinically-translatable percutaneous transendocardial-injection. Three months post-treatment, echocardiography indicated a significant improvement in systolic and diastolic cardiac function. Moreover, improvement in bipolar voltage and decrease in transmural infarct progression was demonstrated by electromechanical NOGA-mapping. Functional benefit was associated with an increase in myocardial vascularization and remodeling. These findings in a large animal model of ischemia-reperfusion demonstrate the feasibility and efficacy of using MPs as a delivery system for growth factors and provide strong evidence to move forward with clinical studies using therapeutic proteins combined with catheter-compatible biomaterials.
Collapse
Affiliation(s)
- Elisa Garbayo
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Juan José Gavira
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Hematology, Cardiology and Cell Therapy, Clínica Universidad de Navarra and Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Manuel Garcia de Yebenes
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Hematology, Cardiology and Cell Therapy, Clínica Universidad de Navarra and Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Beatriz Pelacho
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Hematology, Cardiology and Cell Therapy, Clínica Universidad de Navarra and Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Gloria Abizanda
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Hematology, Cardiology and Cell Therapy, Clínica Universidad de Navarra and Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Hugo Lana
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - María José Blanco-Prieto
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Felipe Prosper
- Instituto de Investigacion Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Hematology, Cardiology and Cell Therapy, Clínica Universidad de Navarra and Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| |
Collapse
|
24
|
Peña B, Martinelli V, Jeong M, Bosi S, Lapasin R, Taylor MG, Long CS, Shandas R, Park D, Mestroni L. Biomimetic Polymers for Cardiac Tissue Engineering. Biomacromolecules 2016; 17:1593-601. [PMID: 27073119 PMCID: PMC4863197 DOI: 10.1021/acs.biomac.5b01734] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/24/2016] [Indexed: 12/26/2022]
Abstract
Heart failure is a morbid disorder characterized by progressive cardiomyocyte (CM) dysfunction and death. Interest in cell-based therapies is growing, but sustainability of injected CMs remains a challenge. To mitigate this, we developed an injectable biomimetic Reverse Thermal Gel (RTG) specifically engineered to support long-term CM survival. This RTG biopolymer provided a solution-based delivery vehicle of CMs, which transitioned to a gel-based matrix shortly after reaching body temperature. In this study we tested the suitability of this biopolymer to sustain CM viability. The RTG was biomolecule-functionalized with poly-l-lysine or laminin. Neonatal rat ventricular myocytes (NRVM) and adult rat ventricular myocytes (ARVM) were cultured in plain-RTG and biomolecule-functionalized-RTG both under 3-dimensional (3D) conditions. Traditional 2D biomolecule-coated dishes were used as controls. We found that the RTG-lysine stimulated NRVM to spread and form heart-like functional syncytia. Regarding cell contraction, in both RTG and RTG-lysine, beating cells were recorded after 21 days. Additionally, more than 50% (p value < 0.05; n = 5) viable ARVMs, characterized by a well-defined cardiac phenotype represented by sarcomeric cross-striations, were found in the RTG-laminin after 8 days. These results exhibit the tremendous potential of a minimally invasive CM transplantation through our designed RTG-cell therapy platform.
Collapse
Affiliation(s)
- Brisa Peña
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| | | | - Mark Jeong
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| | - Susanna Bosi
- I.C.G.E.B. and University
of Trieste, Trieste Italy
| | | | - Matthew
R. G. Taylor
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| | - Carlin S. Long
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| | - Robin Shandas
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| | - Daewon Park
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| | - Luisa Mestroni
- Cardiovascular Institute and Bioengineering Department, University of Colorado−Denver, Aurora, Colorado, United States
| |
Collapse
|
25
|
Onoshima D, Yukawa H, Baba Y. Multifunctional quantum dots-based cancer diagnostics and stem cell therapeutics for regenerative medicine. Adv Drug Deliv Rev 2015; 95:2-14. [PMID: 26344675 DOI: 10.1016/j.addr.2015.08.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/31/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022]
Abstract
A field of recent diagnostics and therapeutics has been advanced with quantum dots (QDs). QDs have developed into new formats of biomolecular sensing to push the limits of detection in biology and medicine. QDs can be also utilized as bio-probes or labels for biological imaging of living cells and tissues. More recently, QDs has been demonstrated to construct a multifunctional nanoplatform, where the QDs serve not only as an imaging agent, but also a nanoscaffold for diagnostic and therapeutic modalities. This review highlights the promising applications of multi-functionalized QDs as advanced nanosensors for diagnosing cancer and as innovative fluorescence probes for in vitro or in vivo stem cell imaging in regenerative medicine.
Collapse
|
26
|
Cardiac atrial appendage stem cells engraft and differentiate into cardiomyocytes in vivo: A new tool for cardiac repair after MI. Int J Cardiol 2015; 201:10-9. [DOI: 10.1016/j.ijcard.2015.07.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/16/2015] [Accepted: 07/26/2015] [Indexed: 12/22/2022]
|
27
|
Suarez SL, Rane AA, Muñoz A, Wright AT, Zhang SX, Braden RL, Almutairi A, McCulloch AD, Christman KL. Intramyocardial injection of hydrogel with high interstitial spread does not impact action potential propagation. Acta Biomater 2015; 26:13-22. [PMID: 26265060 DOI: 10.1016/j.actbio.2015.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/29/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022]
Abstract
Injectable biomaterials have been evaluated as potential new therapies for myocardial infarction (MI) and heart failure. These materials have improved left ventricular (LV) geometry and ejection fraction, yet there remain concerns that biomaterial injection may create a substrate for arrhythmia. Since studies of this risk are lacking, we utilized optical mapping to assess the effects of biomaterial injection and interstitial spread on cardiac electrophysiology. Healthy and infarcted rat hearts were injected with a model poly(ethylene glycol) hydrogel with varying degrees of interstitial spread. Activation maps demonstrated delayed propagation of action potentials across the LV epicardium in the hydrogel-injected group when compared to saline and no-injection groups. However, the degree of the electrophysiological changes depended on the spread characteristics of the hydrogel, such that hearts injected with highly spread hydrogels showed no conduction abnormalities. Conversely, the results of this study indicate that injection of a hydrogel exhibiting minimal interstitial spread may create a substrate for arrhythmia shortly after injection by causing LV activation delays and reducing gap junction density at the site of injection. Thus, this work establishes site of delivery and interstitial spread characteristics as important factors in the future design and use of biomaterial therapies for MI treatment. STATEMENT OF SIGNIFICANCE Biomaterials for treating myocardial infarction have become an increasingly popular area of research. Within the past few years, this work has transitioned to some large animals models, and Phase I & II clinical trials. While these materials have preserved/improved cardiac function the effect of these materials on arrhythmogenesis, which is of considerable concern when injecting anything into the heart, has yet to be understood. Our manuscript is therefore a first of its kind in that it directly examines the potential of an injectable material to create a substrate for arrhythmias. This work suggests that site of delivery and distribution in the tissue are important criteria in the design and development of future biomaterial therapies for myocardial infarction treatment.
Collapse
Affiliation(s)
- Sophia L Suarez
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Aboli A Rane
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Adam Muñoz
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Adam T Wright
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shirley X Zhang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Rebecca L Braden
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Adah Almutairi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA.
| | - Andrew D McCulloch
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karen L Christman
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
28
|
Holt-Casper D, Theisen JM, Moreno AP, Warren M, Silva F, Grainger DW, Bull DA, Patel AN. Novel xeno-free human heart matrix-derived three-dimensional scaffolds. J Transl Med 2015; 13:194. [PMID: 26084398 PMCID: PMC4505384 DOI: 10.1186/s12967-015-0559-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/02/2015] [Indexed: 12/05/2022] Open
Abstract
Rationale Myocardial infarction (MI) results in damaged heart tissue which can progress to severely reduce cardiac function, leading to death. Recent studies have injected dissociated, suspended cardiac cells into coronary arteries to restore function with limited results attributed to poor cell retention and cell death. Extracellular matrix (ECM) injected into damaged cardiac tissue sites show some promising effects. However, combined use of human cardiac ECM and cardiac cells may produce superior benefits to restore cardiac function. Objective This study was designed to assess use of new three-dimensional human heart ECM-derived scaffolds to serve as vehicles to deliver cardiac-derived cells directly to damaged heart tissue and improve cell retention at these sites while also providing biomechanical support and attracting host cell recruitment. Methods and Results ECM-derived porous protein scaffolds were fabricated from human heart tissues. These scaffolds were designed to carry, actively promote and preserve cardiac cell phenotype, viability and functional retention in tissue sites. ECM scaffolds were optimized and were seeded with human cardiomyocytes, cultured and subsequently implanted ex vivo onto infarcted murine epicardium. Seeded human cardiomyocytes readily adhered to human cardiac-derived ECM scaffolds and maintained representative phenotypes including expression of cardiomyocyte-specific markers, and remained electrically synchronous within the scaffold in vitro. Ex vivo, cardiomyocyte-seeded ECM scaffolds spontaneously adhered and incorporated into murine ventricle. Conclusions Decellularized human cardiac tissue-derived 3D ECM scaffolds are effective delivery vehicles for human cardiac cells to directly target ischemic heart tissue and warrant further studies to assess their therapeutic potential in restoring essential cardiac functions. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0559-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dolly Holt-Casper
- Division of Cardiothoracic Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, 84112, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Jeff M Theisen
- Division of Cardiothoracic Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, 84112, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Alonso P Moreno
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, 84112-5000, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Mark Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, 84112-5000, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Francisco Silva
- Division of Cardiothoracic Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, 84112, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - David W Grainger
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, 84112-5000, USA. .,Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - David A Bull
- Division of Cardiothoracic Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, 84112, USA. .,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, 84112-5000, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Amit N Patel
- Division of Cardiothoracic Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, 84112, USA. .,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, 84112-5000, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA. .,University of Utah, 30 N 1900 E SOM 3c127, Salt Lake City, UT, 84132, USA.
| |
Collapse
|
29
|
Pätilä T, Miyagawa S, Imanishi Y, Fukushima S, Siltanen A, Mervaala E, Kankuri E, Harjula A, Sawa Y. Comparison of arrhythmogenicity and proinflammatory activity induced by intramyocardial or epicardial myoblast sheet delivery in a rat model of ischemic heart failure. PLoS One 2015; 10:e0123963. [PMID: 25860790 PMCID: PMC4393220 DOI: 10.1371/journal.pone.0123963] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 03/08/2015] [Indexed: 01/14/2023] Open
Abstract
Although cell therapy of the failing heart by intramyocardial injections of myoblasts to results in regenerative benefit, it has also been associated with undesired and prospectively fatal arrhythmias. We hypothesized that intramyocardial injections of myoblasts could enhance inflammatory reactivity and facilitate electrical cardiac abnormalities that can be reduced by epicardial myoblast sheet delivery. In a rat model of ischemic heart failure, myoblast therapy either by intramyocardial injections or epicardial cell sheets was given 2 weeks after occlusion of the coronary artery. Ventricular premature contractions (VPCs) were assessed, using an implanted three-lead electrocardiograph at 1, 7, and 14 days after therapy, and 16-point epicardial electropotential mapping (EEPM) was used to evaluate ventricular arrhythmogenicity under isoproterenol stress. Cardiac functioning was assessed by echocardiography. Both transplantation groups showed therapeutic benefit over sham therapy. However, VPCs were more frequent in the Injection group on day 1 and day 14 after therapy than in animals receiving epicardial or sham therapy (p < 0.05 and p < 0.01, respectively). EEPM under isoproterenol stress showed macroreentry at the infarct border area, leading to ventricular tachycardias in the Injection group, but not in the myoblast sheet- or sham-treated groups (p = 0.045). Both transplantation types modified the myocardial cytokine expression profile. In animals receiving epicardial myoblast therapy, selective reductions in the expressions of interferon gamma, interleukin (IL)-1β and IL12 were observed, accompanied by reduced infiltration of inflammatory CD11b- and CD68-positive leukocytes, compared with animals receiving myoblasts as intramyocardial injections. Intramyocardial myoblast delivery was associated with enhanced inflammatory and immunomodulatory reactivity and increased frequency of VPCs. In comparison to intramyocardial injection, the epicardial route may serve as the preferred method of skeletal myoblast transplantation to treat heart failure.
Collapse
Affiliation(s)
- Tommi Pätilä
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pediatric Cardiac Surgery, Hospital for Children and Adolescents, University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Shigeru Miyagawa
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yukiko Imanishi
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satsuki Fukushima
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Eero Mervaala
- Pharmacology, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Pharmacology, University of Helsinki, Helsinki, Finland
| | - Ari Harjula
- Department of Cardiothoracic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Yoshiki Sawa
- Department of Cardiothoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
30
|
Almeida SO, Skelton RJ, Adigopula S, Ardehali R. Arrhythmia in stem cell transplantation. Card Electrophysiol Clin 2015; 7:357-70. [PMID: 26002399 DOI: 10.1016/j.ccep.2015.03.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Stem cell regenerative therapies hold promise for treating diseases across the spectrum of medicine. While significant progress has been made in the preclinical stages, the clinical application of cardiac cell therapy is limited by technical challenges. Certain methods of cell delivery, such as intramyocardial injection, carry a higher rate of arrhythmias. Other potential contributors to the arrhythmogenicity of cell transplantation include reentrant pathways caused by heterogeneity in conduction velocities between graft and host as well as graft automaticity. In this article, the arrhythmogenic potential of cell delivery to the heart is discussed.
Collapse
Affiliation(s)
- Shone O Almeida
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA
| | - Rhys J Skelton
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA; Murdoch Children's Research Institute, The Royal Children's Hospital, Cardiac Development, 50 Flemington Road, Parkville, Victoria 3052, Australia
| | - Sasikanth Adigopula
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 630 East, Los Angeles, CA 90095, USA; Eli and Edyth Broad Stem Cell Research Center, University of California, 675 Charles E Young Drive South, MRL Room 3780, Los Angeles, CA 90095, USA.
| |
Collapse
|
31
|
Oommen S, Yamada S, Cantero Peral S, Campbell KA, Bruinsma ES, Terzic A, Nelson TJ. Human umbilical cord blood-derived mononuclear cells improve murine ventricular function upon intramyocardial delivery in right ventricular chronic pressure overload. Stem Cell Res Ther 2015; 6:50. [PMID: 25890300 PMCID: PMC4416353 DOI: 10.1186/s13287-015-0044-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 10/17/2014] [Accepted: 03/05/2015] [Indexed: 02/03/2023] Open
Abstract
Introduction Stem cell therapy has emerged as potential therapeutic strategy for damaged heart muscles. Umbilical cord blood (UCB) cells are the most prevalent stem cell source available, yet have not been fully tested in cardiac regeneration. Herein, studies were performed to evaluate the cardiovascular safety and beneficial effect of mononuclear cells (MNCs) isolated from human umbilical cord blood upon intramyocardial delivery in a murine model of right ventricle (RV) heart failure due to pressure overload. Methods UCB-derived MNCs were delivered into the myocardium of a diseased RV cardiac model. Pulmonary artery banding (PAB) was used to produce pressure overload in athymic nude mice that were then injected intramyocardially with UCB-MNCs (0.4 × 10^6 cells/heart). Cardiac functions were then monitored by telemetry, echocardiography, magnetic resonance imaging (MRI) and pathologic analysis of heart samples to determine the ability for cell-based repair. Results The cardio-toxicity studies provided evidence that UCB cell transplantation has a safe therapeutic window between 0.4 to 0.8 million cells/heart without altering QT or ST-segments or the morphology of electrocardiograph waves. The PAB cohort demonstrated significant changes in RV chamber dilation and functional defects consistent with severe pressure overload. Using cardiac MRI analysis, UCB-MNC transplantation in the setting of PAB demonstrated an improvement in RV structure and function in this surgical mouse model. The RV volume load in PAB-only mice was 24.09 ± 3.9 compared to 11.05 ± 2.09 in the cell group (mm3, P-value <0.005). The analysis of pathogenic gene expression (BNP, ANP, Acta1, Myh7) in the cell-transplanted group showed a significant reversal with respect to the diseased PAB mice with a robust increase in cardiac progenitor gene expression such as GATA4, Kdr, Mef2c and Nkx2.5. Histological analysis indicated significant fibrosis in the RV in response to PAB that was reduced following UCB-MNC’s transplantation along with concomitant increased Ki-67 expression and CD31 positive vessels as a marker of angiogenesis within the myocardium. Conclusions These findings indicate that human UCB-derived MNCs promote an adaptive regenerative response in the right ventricle upon intramyocardial transplantation in the setting of chronic pressure overload heart failure.
Collapse
Affiliation(s)
- Saji Oommen
- General Internal Medicine and Transplant Center, Mayo Clinic, Rochester, MN, USA. .,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| | - Satsuki Yamada
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| | - Susana Cantero Peral
- General Internal Medicine and Transplant Center, Mayo Clinic, Rochester, MN, USA. .,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Autonomous University of Barcelona, Program of Doctorate of Internal Medicine, Barcelona, Spain.
| | - Katherine A Campbell
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| | - Elizabeth S Bruinsma
- General Internal Medicine and Transplant Center, Mayo Clinic, Rochester, MN, USA.
| | - Andre Terzic
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA. .,Department of Medical Genetics, Mayo Clinic, Rochester, MN, USA.
| | - Timothy J Nelson
- General Internal Medicine and Transplant Center, Mayo Clinic, Rochester, MN, USA. .,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA. .,Department of Medicine, Mayo Clinic, 200 First Street, SW, Rochester, MN, 55905, USA.
| |
Collapse
|
32
|
Tung W, Weintraub NL, Berman AE, Tang Y. A novel high throughput approach to screen for cardiac arrhythmic events following stem cell treatment. Med Hypotheses 2015; 84:294-7. [PMID: 25655221 DOI: 10.1016/j.mehy.2015.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/11/2015] [Indexed: 12/19/2022]
Abstract
Although stem cell therapy is promising for repairing damaged cardiac tissue and improving heart function, there are safety concerns, especially regarding the risk of arrhythmias, which can be life threatening. To address this issue, we propose to develop a novel screening system to evaluate arrhythmic risk associated with stem cell therapy using a high-throughput multielectrode array system that can measure conduction velocity and action potential duration in cardiomyocytes co-cultured with different types of stem cells, such as mesenchymal stem cells, skeletal myoblasts, and resident cardiac stem cells. We will assess the arrhythmic potential of each of these types of stem cells under normoxic and hypoxic conditions, with/without application of oxidative stress or catecholamines. We hypothesize that these methods will prove to be an effective way to screen for arrhythmic risk of cardiac stem cell therapy. Ultimately, our approach can potentially be personalized to develop a robust screening protocol in order to identify which stem cell type carries the least amount of risk for arrhythmia. This system will have great clinical benefit to improve the risk/benefit ratio of human stem cell therapy for heart disease.
Collapse
Affiliation(s)
- William Tung
- Vascular Biology Center, Department of Medicine, Medical College of Georgia/Georgia Regents University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Department of Medicine, Medical College of Georgia/Georgia Regents University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Adam E Berman
- Vascular Biology Center, Department of Medicine, Medical College of Georgia/Georgia Regents University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Yaoliang Tang
- Vascular Biology Center, Department of Medicine, Medical College of Georgia/Georgia Regents University, 1459 Laney Walker Blvd, Augusta, GA 30912, USA.
| |
Collapse
|
33
|
Antanavičiūtė I, Ereminienė E, Vysockas V, Račkauskas M, Skipskis V, Rysevaitė K, Treinys R, Benetis R, Jurevičius J, Skeberdis VA. Exogenous connexin43-expressing autologous skeletal myoblasts ameliorate mechanical function and electrical activity of the rabbit heart after experimental infarction. Int J Exp Pathol 2014; 96:42-53. [PMID: 25529770 DOI: 10.1111/iep.12109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 10/26/2014] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction is one of the major causes of mortality worldwide. For regeneration of the rabbit heart after experimentally induced infarction we used autologous skeletal myoblasts (SMs) due to their high proliferative potential, resistance to ischaemia and absence of immunological and ethical concerns. The cells were characterized with muscle-specific and myogenic markers. Cell transplantation was performed by injection of cell suspension (0.5 ml) containing approximately 6 million myoblasts into the infarction zone. The animals were divided into four groups: (i) no injection; (ii) sham injected; (iii) injected with wild-type SMs; and (iv) injected with SMs expressing connexin43 fused with green fluorescent protein (Cx43EGFP). Left ventricular ejection fraction (LVEF) was evaluated by 2D echocardiography in vivo before infarction, when myocardium has stabilized after infarction, and 3 months after infarction. Electrical activity in the healthy and infarction zones of the heart was examined ex vivo in Langendorff-perfused hearts by optical mapping using di-4-ANEPPS, a potential sensitive fluorescent dye. We demonstrate that SMs in the coculture can couple electrically not only to abutted but also to remote acutely isolated allogenic cardiac myocytes through membranous tunnelling tubes. The beneficial effect of cellular therapy on LVEF and electrical activity was observed in the group of animals injected with Cx43EGFP-expressing SMs. L-type Ca(2+) current amplitude was approximately fivefold smaller in the isolated SMs compared to healthy myocytes suggesting that limited recovery of LVEF may be related to inadequate expression or function of L-type Ca(2+) channels in transplanted differentiating SMs.
Collapse
Affiliation(s)
- Ieva Antanavičiūtė
- Institute of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang H, Wang Z, Jiang H, Ma D, Zhou W, Zhang G, Chen W, Huang J, Liu Y. Effect of autologous bone marrow cell transplantation combined with off-pump coronary artery bypass grafting on cardiac function in patients with chronic myocardial infarction. Cardiology 2014; 130:27-33. [PMID: 25501100 DOI: 10.1159/000369381] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 10/24/2014] [Indexed: 11/19/2022]
Abstract
OBJECTIVES This study aimed to investigate the feasibility and effects of intramuscular injections of autologous bone marrow cells (BMC) combined with off-pump coronary artery bypass grafts (OPCAB) on improving cardiac function in chronic myocardial infarction patients. METHODS Ninety patients with chronic myocardial infarction were prospectively enrolled and randomized to an OPCAB with saline or an OPCAB with BMC-treatment group. After finishing CABG, patients received injections of BMC or saline into the marginal area of the infarct. The primary endpoint was incidence of emergent adverse events within 6 months. RESULTS There were no differences between the control and BMC-treated groups in baseline ejection fractions (EF) or wall motion score indices (WMSI) in the affected segments. At the 6-month follow-up, the ejection fraction was significantly increased in the BMC-treated group compared to controls (47.58 ± 6.34 vs. 40.11 ± 7.42; p < 0.05), whereas the WMSI were significantly decreased (1.25 ± 0.32 vs. 1.54 ± 0.53; p < 0.05), with no occurrences of life-threatening arrhythmias or death. The addition of BMC injections to OPCAB treatment increased regional perfusion to the marginal infarct area. CONCLUSION These results demonstrate that BMC transplant is beneficial to the cardiac function with no adverse effects, and therefore a safe and feasible adjunct therapy providing beneficial effects in clinical practice.
Collapse
Affiliation(s)
- Huishan Wang
- Department of Cardiac Surgery, Shenyang Northern Hospital, Shenyang, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Right ventricular failure secondary to chronic overload in congenital heart diseases: benefits of cell therapy using human embryonic stem cell-derived cardiac progenitors. J Thorac Cardiovasc Surg 2014; 149:708-15.e1. [PMID: 25583108 DOI: 10.1016/j.jtcvs.2014.11.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/07/2014] [Accepted: 11/16/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Despite the increasing incidence of right ventricular (RV) failure in adult patients with congenital heart disease, current therapeutic options are still limited. By contrast to left-heart diseases, cell-based myocardial regeneration applied to the right ventricle is poorly studied, even though it may be a therapeutic solution. As human embryonic stem cell-derived cardiac progenitors seem to be good candidates owing to their proliferation capacity, our aim was to assess, in a large animal model of overloaded RV dysfunction, the feasibility and effects of such a cell therapy. METHODS Human MesP1(+)/SSEA-1(+) cardiogenic mesodermal cells were administered using multiple intramyocardial injections 4 months after a surgical procedure mimicking the repaired tetralogy of Fallot, and their effects were observed 3 months later on hemodynamic, rhythmic, and histologic parameters. RESULTS All pigs (sham n = 6, treated n = 6) survived without complication, and cell therapy was clinically well tolerated. Although functional, contractility, and energetics parameters evolved similarly in both groups, benefits regarding arrhythmic susceptibility were observed in the treated group, associated with a significant decrease of peri-myocyte fibrosis (5.71% ± 2.49% vs 12.12% ± 1.85%; P < .01) without interstitial fibrosis change (5.18% ± 0.81% vs 5.49% ± 1.01%). Such a decrease could be related to paracrine effects, as no human cells could be detected within the myocardium. CONCLUSIONS Cell therapy using intramyocardial injections of human MesP1(+)/SSEA-1(+) cardiogenic mesodermal cells seems to have benefits regarding overloaded RV tissue remodeling and arrhythmic susceptibility, but this mode of administration is not sufficient to obtain a significant improvement in RV function.
Collapse
|
36
|
Hyperthermia differently affects connexin43 expression and gap junction permeability in skeletal myoblasts and HeLa cells. Mediators Inflamm 2014; 2014:748290. [PMID: 25143668 PMCID: PMC4131114 DOI: 10.1155/2014/748290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 12/11/2022] Open
Abstract
Stress kinases can be activated by hyperthermia and modify the expression level and properties of membranous and intercellular channels. We examined the role of c-Jun NH2-terminal kinase (JNK) in hyperthermia-induced changes of connexin43 (Cx43) expression and permeability of Cx43 gap junctions (GJs) in the rabbit skeletal myoblasts (SkMs) and Cx43-EGFP transfected HeLa cells. Hyperthermia (42°C for 6 h) enhanced the activity of JNK and its target, the transcription factor c-Jun, in both SkMs and HeLa cells. In SkMs, hyperthermia caused a 3.2-fold increase in the total Cx43 protein level and enhanced the efficacy of GJ intercellular communication (GJIC). In striking contrast, hyperthermia reduced the total amount of Cx43 protein, the number of Cx43 channels in GJ plaques, the density of hemichannels in the cell membranes, and the efficiency of GJIC in HeLa cells. Both in SkMs and HeLa cells, these changes could be prevented by XG-102, a JNK inhibitor. In HeLa cells, the changes in Cx43 expression and GJIC under hyperthermic conditions were accompanied by JNK-dependent disorganization of actin cytoskeleton stress fibers while in SkMs, the actin cytoskeleton remained intact. These findings provide an attractive model to identify the regulatory players within signalosomes, which determine the cell-dependent outcomes of hyperthermia.
Collapse
|
37
|
Pavo N, Charwat S, Nyolczas N, Jakab A, Murlasits Z, Bergler-Klein J, Nikfardjam M, Benedek I, Benedek T, Pavo IJ, Gersh BJ, Huber K, Maurer G, Gyöngyösi M. Cell therapy for human ischemic heart diseases: critical review and summary of the clinical experiences. J Mol Cell Cardiol 2014; 75:12-24. [PMID: 24998410 DOI: 10.1016/j.yjmcc.2014.06.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/23/2014] [Accepted: 06/26/2014] [Indexed: 12/24/2022]
Abstract
A decade ago, stem or progenitor cells held the promise of tissue regeneration in human myocardium, with the expectation that these therapies could rescue ischemic myocyte damage, enhance vascular density and rebuild injured myocardium. The accumulated evidence in 2014 indicates, however, that the therapeutic success of these cells is modest and the tissue regeneration involves much more complex processes than cell-related biologics. As the quest for the ideal cell or combination of cells continues, alternative cell types, such as resident cardiac cells, adipose-derived or phenotypic modified stem or progenitor cells have also been applied, with the objective of increasing both the number and the retention of the reparative cells in the myocardium. Two main delivery routes (intracoronary and percutaneous intramyocardial) of stem cells are currently used preferably for patients with recent acute myocardial infarction or ischemic cardiomyopathy. Other delivery modes, such as surgical or intravenous via peripheral veins or coronary sinus have also been utilized with less success. Due to the difficult recruitment of patients within conceivable timeframe into cardiac regenerative trials, meta-analyses of human cardiac cell-based studies have tried to gather sufficient number of subjects to present a statistical compelling statement, reporting modest success with a mean increase of 0.9-6.1% in left ventricular global ejection fraction. Additionally, nearly half of the long-term studies reported the disappearance of the initial benefit of this treatment. Beside further extensive efforts to increase the efficacy of currently available methods, pre-clinical experiments using new techniques such as tissue engineering or exploiting paracrine effect hold promise to regenerate injured human cardiac tissue.
Collapse
Affiliation(s)
- Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Austria
| | - Silvia Charwat
- Department of Cardiology, Medical University of Vienna, Austria
| | - Noemi Nyolczas
- Department of Cardiology, Medical University of Vienna, Austria
| | - András Jakab
- Department of Biomedical Laboratory and Imaging Science, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Murlasits
- Exercise Biochemistry Laboratory, The University of Memphis, Department of Health and Sport Sciences, Memphis, TN, USA
| | | | | | - Imre Benedek
- Department of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Romania
| | - Teodora Benedek
- Department of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Romania
| | - Imre J Pavo
- Department of Cardiology, Medical University of Vienna, Austria
| | - Bernard J Gersh
- Internal Medicine, Mayo Graduate School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kurt Huber
- 3(rd) Dept. Cardiology and Emergency Medicine, Wilhelminen hospital, Vienna, Austria
| | - Gerald Maurer
- Department of Cardiology, Medical University of Vienna, Austria
| | | |
Collapse
|
38
|
de Souza ASB, Souza WKSB, Costa SA, Freitas EMDM, Carvalho G, Sá LAB, Rassi S. Incidence of ventricular arrhythmias after stem cell therapy in patients with Chagas cardiomyopathy. Arq Bras Cardiol 2014; 102:489-94. [PMID: 24918914 PMCID: PMC4051452 DOI: 10.5935/abc.20140053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 12/18/2013] [Indexed: 11/20/2022] Open
Abstract
Background Treatment with stem cells in several cardiomyopathies may be related to the
increase in arrhythmias. Objectives To determine whether intracoronary injection of stem cells in patients with Chagas
cardiomyopathy is associated with increased incidence of ventricular arrhythmias,
compared to the Control Group. Methods A retrospective cohort study that evaluated the medical records of 60 patients who
participated in a previous cross-sectional study. The following data were
collected: age, gender, drugs used and Holter variables that demonstrated the
presence of arrhythmias. Holter was performed in four stages: randomization, 2, 6
and 12 months segments. The Control Group received medical treatment and
intracoronary injection of placebo and the Study Group had drug treatment and
autologous stem cell implant. Results There was no difference between Control Group and Study Group when analyzing the
arrhythmia criteria. In the intra-group analysis, significant difference was found
between the Holter tests of the Study Group for the variable total ventricular
premature beats when compared with baseline, with p = 0.014 between Holter at
randomization and Holter at 2 months, p = 0.004 between Holter at randomization
and Holter at 6 months, and p = 0.014 between Holter at randomization and Holter
at 12 months. The variable non-sustained ventricular tachycardia between Holter at
randomization and Holter at 6 months showed p = 0.036. Conclusion The intracoronary injection of stem cells did not increase the incidence of
ventricular arrhythmias in patients with Chagas cardiomyopathy compared to the
Control Group.
Collapse
Affiliation(s)
- Adriana Sebba Barroso de Souza
- Mailing Address: Adriana Sebba Barroso de Souza, Universidade Federal de
Goiás, - Av. Esperança, s/n - Setor Itatiaia. Postal Code 74001-970, Goiânia, GO -
Brazil. E-mail:
| | | | | | | | | | | | | |
Collapse
|
39
|
Emmert MY, Hitchcock RW, Hoerstrup SP. Cell therapy, 3D culture systems and tissue engineering for cardiac regeneration. Adv Drug Deliv Rev 2014; 69-70:254-69. [PMID: 24378579 DOI: 10.1016/j.addr.2013.12.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/06/2013] [Accepted: 12/17/2013] [Indexed: 01/02/2023]
Abstract
Ischemic Heart Disease (IHD) still represents the "Number One Killer" worldwide accounting for the death of numerous patients. However the capacity for self-regeneration of the adult heart is very limited and the loss of cardiomyocytes in the infarcted heart leads to continuous adverse cardiac-remodeling which often leads to heart-failure (HF). The concept of regenerative medicine comprising cell-based therapies, bio-engineering technologies and hybrid solutions has been proposed as a promising next-generation approach to address IHD and HF. Numerous strategies are under investigation evaluating the potential of regenerative medicine on the failing myocardium including classical cell-therapy concepts, three-dimensional culture techniques and tissue-engineering approaches. While most of these regenerative strategies have shown great potential in experimental studies, the translation into a clinical setting has either been limited or too rapid leaving many key questions unanswered. This review summarizes the current state-of-the-art, important challenges and future research directions as to regenerative approaches addressing IHD and resulting HF.
Collapse
|
40
|
Zheng SX, Weng YL, Zhou CQ, Wen ZZ, Huang H, Wu W, Wang JF, Wang T. Comparison of cardiac stem cells and mesenchymal stem cells transplantation on the cardiac electrophysiology in rats with myocardial infarction. Stem Cell Rev Rep 2014; 9:339-49. [PMID: 22544360 DOI: 10.1007/s12015-012-9367-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Whether transplanted cardiac stem cells (CSCs) and mesenchymal stem cells (MSCs) improved ventricular fibrillation threshold (VFT) similarly is still unclear. We sought to compare the effects of the CSC and MSC transplantation on the electrophysiological characteristics and VFT in rats with myocardial infarction (MI). METHODS MI was induced in 30 male Sprague-Dawley rats. Two weeks later, animals were randomized to receive 5 × 10(6) CSCs labeled with PKH26 in PBS or 5 × 10(6) MSCs labeled with PKH26 in phosphate buffer solution(PBS) or PBS alone injection into the infarcted anterior ventricular free wall. Six weeks after the injection, electrophysiological characteristics and VFT were measured. Labeled CSCs and MSCs were observed in 5 μm cryostat sections from each heart. RESULTS Malignant ventricular arrhythmias were significantly (P = 0.0055) less inducible in the CSC group than the MSC group. The VFTs were improved in the CSC group compared with the MSC group. Labeled CSCs and MSCs were identified in the infarct zone and infarct marginal zone. Labeled CSCs expressed Connexin-43, von Willebrand factor, α-smooth muscle actin and α-sarcomeric actin,while the Labeled MSCs expressed von Willebrand factor, α-smooth muscle actin and α-sarcomeric actin in vivo. CONCLUSIONS After 6 weeks of cell transplantation, CSCs are superior to MSCs in modulating the electrophysiological abnormality and improving the VFT in rats with MI. CSCs and MSCs express markers that suggest muscle, endothelium and vascular smooth muscle phenotypes in vivo, but MSCs rarely express Connexin-43.
Collapse
Affiliation(s)
- Shao-Xin Zheng
- Cardiovascular Medicine, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, 107 Yanjiang Xi Road, Guangzhou, 510120, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Liao SY, Tse HF. Multipotent (adult) and pluripotent stem cells for heart regeneration: what are the pros and cons? Stem Cell Res Ther 2013; 4:151. [PMID: 24476362 PMCID: PMC4056686 DOI: 10.1186/scrt381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heart failure after myocardial infarction is the leading cause of mortality and morbidity worldwide. Existing medical and interventional therapies can only reduce the loss of cardiomyocytes during myocardial infarction but are unable to replenish the permanent loss of cardiomyocytes after the insult, which contributes to progressive pathological left ventricular remodeling and progressive heart failure. As a result, cell-based therapies using multipotent (adult) stem cells and pluripotent stem cells (embryonic stem cells or induced pluripotent stem cells) have been explored as potential therapeutic approaches to restore cardiac function in heart failure. Nevertheless, the optimal cell type with the best therapeutic efficacy and safety for heart regeneration is still unknown. In this review, the potential pros and cons of different types of multipotent (adult) stem cells and pluripotent stem cells that have been investigated in preclinical and clinical studies are reviewed, and the future perspective of stem cell-based therapy for heart regeneration is discussed.
Collapse
|
42
|
Martin I, Baldomero H, Bocelli-Tyndall C, Emmert MY, Hoerstrup SP, Ireland H, Passweg J, Tyndall A. The survey on cellular and engineered tissue therapies in Europe in 2011. Tissue Eng Part A 2013; 20:842-53. [PMID: 24090467 DOI: 10.1089/ten.tea.2013.0372] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Following the coordinated efforts of five established scientific organizations, this report describes the "novel cellular therapy" activity (i.e., cellular treatments excluding hematopoietic stem cells [HSC] for the reconstitution of hematopoiesis) in Europe for the year 2011. Two hundred forty-six teams from 35 countries responded to the cellular therapy survey, 126 teams from 24 countries provided data on 1759 patients using a dedicated survey and 120 teams reported no activity. Indications were musculoskeletal/rheumatological disorders (46%; 99% autologous), cardiovascular disorders (22%; 100% autologous), hematology/oncology, predominantly including the prevention or treatment of graft-versus-host disease (18%; 2% autologous), neurological disorders (2%; 83% autologous), gastrointestinal (1%; 68% autologous), and other indications (12%; 77% autologous). Autologous cells were used predominantly for musculoskeletal/rheumatological (58%) and cardiovascular (27%) disorders, whereas allogeneic cells were used mainly for hematology/oncology (84%). The reported cell types were mesenchymal stem/stromal cells (56%), HSC (23%), chondrocytes (12%), dermal fibroblasts (3%), keratinocytes (2%), and others (4%). In 40% of the grafts, cells were delivered following ex vivo expansion, whereas cells were transduced or sorted, respectively, in 3% and 10% of the reported cases. Cells were delivered intraorgan (42%), intravenously (26%), on a membrane or gel (16%), or using 3D scaffolds (16%). Compared to last year, the number of teams participating in the dedicated survey doubled and, for the first time, all European Group for Blood and Marrow Transplantation teams reporting information on cellular therapies completed the extended questionnaire. The data are compared with those collected since 2008 to identify trends in the field. This year's edition specifically focuses on cardiac cell therapy.
Collapse
Affiliation(s)
- Ivan Martin
- 1 Department of Surgery, University Hospital Basel , University of Basel, Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Berry SE, Andruszkiewicz P, Chun JL, Hong J. Nestin expression in end-stage disease in dystrophin-deficient heart: implications for regeneration from endogenous cardiac stem cells. Stem Cells Transl Med 2013; 2:848-61. [PMID: 24068741 PMCID: PMC3808200 DOI: 10.5966/sctm.2012-0174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 05/28/2013] [Indexed: 01/16/2023] Open
Abstract
Nestin(+) cardiac stem cells differentiate into striated cells following myocardial infarct. Transplantation of exogenous stem cells into myocardium of a murine model for Duchenne muscular dystrophy (DMD) increased proliferation of endogenous nestin(+) stem cells and resulted in the appearance of nestin(+) striated cells. This correlated with, and may be responsible for, prevention of dilated cardiomyopathy. We examined nestin(+) stem cells in the myocardium of dystrophin/utrophin-deficient (mdx/utrn(-/-)) mice, a model for DMD. We found that 92% of nestin(+) interstitial cells expressed Flk-1, a marker present on cardiac progenitor cells that differentiate into the cardiac lineage, and that a subset expressed Sca-1, present on adult cardiac cells that become cardiomyocytes. Nestin(+) interstitial cells maintained expression of Flk-1 but lost Sca-1 expression with age and were present in lower numbers in dystrophin-deficient heart than in wild-type heart. Unexpectedly, large clusters of nestin(+) striated cells ranging in size from 20 to 250 cells and extending up to 500 μm were present in mdx/utrn(-/-) heart near the end stage of disease. These cells were also present in dystrophin-deficient mdx/utrn(+/-) and mdx heart but not wild-type heart. Nestin(+) striated cells expressed cardiac troponin I, desmin, and Connexin 43 and correlated with proinflammatory CD68(+) macrophages. Elongated nestin(+) interstitial cells with striations were observed that did not express Flk-1 or the late cardiac marker cardiac troponin I but strongly expressed the early cardiac marker desmin. Nestin was also detected in endothelial and smooth muscle cells. These data indicate that new cardiomyocytes form in dystrophic heart, and nestin(+) interstitial cells may generate them in addition to other cells of the cardiac lineage.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Ly/genetics
- Antigens, Ly/metabolism
- Biomarkers/metabolism
- Connexin 43/genetics
- Connexin 43/metabolism
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Dystrophin/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/physiology
- Heart/physiopathology
- Macrophages/metabolism
- Macrophages/physiology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Nestin/biosynthesis
- Nestin/genetics
- Nestin/metabolism
- Regeneration/genetics
- Regeneration/physiology
- Stem Cells/metabolism
- Stem Cells/physiology
- Utrophin/deficiency
- Utrophin/genetics
- Utrophin/metabolism
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Suzanne E. Berry
- Department of Comparative Biosciences
- Institute for Genomic Biology
- Neuroscience Program, and
| | | | - Ju Lan Chun
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, USA
| | - Jun Hong
- Department of Comparative Biosciences
| |
Collapse
|
44
|
du Pré BC, Doevendans PA, van Laake LW. Stem cells for cardiac repair: an introduction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2013; 10:186-97. [PMID: 23888179 PMCID: PMC3708059 DOI: 10.3969/j.issn.1671-5411.2013.02.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 02/16/2013] [Accepted: 04/22/2013] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease is a major cause of morbidity and mortality throughout the world. Most cardiovascular diseases, such as ischemic heart disease and cardiomyopathy, are associated with loss of functional cardiomyocytes. Unfortunately, the heart has a limited regenerative capacity and is not able to replace these cardiomyocytes once lost. In recent years, stem cells have been put forward as a potential source for cardiac regeneration. Pre-clinical studies that use stem cell-derived cardiac cells show promising results. The mechanisms, though, are not well understood, results have been variable, sometimes transient in the long term, and often without a mechanistic explanation. There are still several major hurdles to be taken. Stem cell-derived cardiac cells should resemble original cardiac cell types and be able to integrate in the damaged heart. Integration requires administration of stem cell-derived cardiac cells at the right time using the right mode of delivery. Once delivered, transplanted cells need vascularization, electrophysiological coupling with the injured heart, and prevention of immunological rejection. Finally, stem cell therapy needs to be safe, reproducible, and affordable. In this review, we will give an introduction to the principles of stem cell based cardiac repair.
Collapse
Affiliation(s)
- Bastiaan C du Pré
- Departments of Cardiology and Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, P.O. box 85500, 3508 GA Utrecht, the Netherlands
| | | | | |
Collapse
|
45
|
Enhanced cardiomyogenic lineage differentiation of adult bone-marrow-derived stem cells grown on cardiogel. Cell Tissue Res 2013; 353:443-56. [PMID: 23771778 DOI: 10.1007/s00441-013-1661-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/06/2013] [Indexed: 02/03/2023]
Abstract
The extracellular matrix (ECM) and its components are known to promote growth and cellular differentiation in vitro. Cardiogel, a three-dimensional extracellular matrix derived from cardiac fibroblasts, is evaluated for its cardiomyogenic-differentiation-inducing potential on bone-marrow-derived stem cells (BMSC). BMSC from adult mice were grown on cardiogel and induced to differentiate into specific lineages that were validated by morphological, phenotypic and molecular assays. The data revealed that the cardiogel enhanced cardiomyogenic and adipogenic differentiation and relegated osteogenic differentiation following specific induction. More importantly, increased cardiomyogenic differentiation was also observed following BMSC growth on cardiogel without specific chemical (5-azacytidine) induction. This is the first report of an attempt to use cardiogel as a biomaterial on which to achieve cardiomyogenic differentiation of BMSC without chemical induction. Our study suggests that cardiogel is an efficient extracellular matrix that enhances the cardiomyogenic differentiation of BMSC and that it can therefore be used as a scaffold for cardiac tissue regeneration.
Collapse
|
46
|
Zhang D, Wang L, Zhang F, Li C, Zhu T, Cao K, Ma W, Yang Z. Nine-year follow-up of local implantation of autologous skeletal myoblasts in a patient with coronary heart disease. AMERICAN JOURNAL OF CASE REPORTS 2013; 14:139-42. [PMID: 23826452 PMCID: PMC3700486 DOI: 10.12659/ajcr.883903] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 02/08/2013] [Indexed: 12/03/2022]
Abstract
Patient: gender – Male, age – 63 year-old Primary Diagnosis: Acute myocardial infarction Co-existing Diseases: Hypertension Medication: Aspirin • beta-blocker • captopril Clinical Procedure: CABG • autologous skeletal myoblast transplantation • PCI Specialty: Cardiology Objective: Unusual or unexpected effect of treatment. Background: Cell transplantation has been viewed as a promising strategy for end-stage heart failure, but long-term follow-up results are lacking. Case Report: In December 2002 we began transplanting autologous skeletal myoblasts in one patient because of serious coronary heart disease. Here, we present the 9-year follow-up results of this patient. No ventricular tachyarrhythmias were detected after treatment. The patient had another myocardial infarction in April 2012 and was treated successful with PCI. Conclusions: Autologous skeletal myoblast transplantation with bypass surgery is associated with improvement in cardiac function and lack of adverse effects in long-term follow-up, making it a promising therapy for patients with heart failure.
Collapse
Affiliation(s)
- Dingguo Zhang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Critical path in cardiac stem cell therapy: an update on cell delivery. Cytotherapy 2013; 15:399-415. [DOI: 10.1016/j.jcyt.2012.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 09/25/2012] [Accepted: 11/02/2012] [Indexed: 01/14/2023]
|
48
|
Sekiya N, Tobita K, Beckman S, Okada M, Gharaibeh B, Sawa Y, Kormos RL, Huard J. Muscle-derived stem cell sheets support pump function and prevent cardiac arrhythmias in a model of chronic myocardial infarction. Mol Ther 2013; 21:662-9. [PMID: 23319053 DOI: 10.1038/mt.2012.266] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Direct intracardiac cell injection for heart repair is hindered by numerous limitations including: cell death, poor spreading of the injected cells, arrhythmia, needle injury, etc. Tissue-engineered cell sheet implantation has the potential to overcome some of these limitations. We evaluated whether the transplantation of a muscle-derived stem cell (MDSC) sheet could improve the regenerative capacity of MDSCs in a chronic model of myocardial infarction. MDSC sheet-implanted mice displayed a reduction in left ventricle (LV) dilation and sustained LV contraction compared with the other groups. The MDSC sheet formed aligned myotubes and produced a significant increase in capillary density and a reduction of myocardial fibrosis compared with the other groups. Hearts transplanted with the MDSC sheets did not display any significant arrhythmias and the donor MDSC survival rate was higher than the direct myocardial MDSC injection group. MDSC sheet implantation yielded better functional recovery of chronic infarcted myocardium without any significant arrhythmic events compared with direct MDSC injection, suggesting this cell sheet delivery system could significantly improve the myocardial regenerative potential of the MDSCs.
Collapse
Affiliation(s)
- Naosumi Sekiya
- Stem Cell Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Minami I, Yamada K, Otsuji TG, Yamamoto T, Shen Y, Otsuka S, Kadota S, Morone N, Barve M, Asai Y, Tenkova-Heuser T, Heuser JE, Uesugi M, Aiba K, Nakatsuji N. A small molecule that promotes cardiac differentiation of human pluripotent stem cells under defined, cytokine- and xeno-free conditions. Cell Rep 2012; 2:1448-60. [PMID: 23103164 DOI: 10.1016/j.celrep.2012.09.015] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/18/2012] [Accepted: 09/12/2012] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including embryonic stem cells and induced pluripotent stem cells, are potentially useful in regenerative therapies for heart disease. For medical applications, clinical-grade cardiac cells must be produced from hPSCs in a defined, cost-effective manner. Cell-based screening led to the discovery of KY02111, a small molecule that promotes differentiation of hPSCs to cardiomyocytes. Although the direct target of KY02111 remains unknown, results of the present study suggest that KY02111 promotes differentiation by inhibiting WNT signaling in hPSCs but in a manner that is distinct from that of previously studied WNT inhibitors. Combined use of KY02111 and WNT signaling modulators produced robust cardiac differentiation of hPSCs in a xeno-free, defined medium, devoid of serum and any kind of recombinant cytokines and hormones, such as BMP4, Activin A, or insulin. The methodology has potential as a means for the practical production of human cardiomyocytes for regeneration therapies.
Collapse
Affiliation(s)
- Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Differentiated adult cardiomyocytes (CMs) lack significant regenerative potential, which is one reason why degenerative heart diseases are the leading cause of death in the western world. For future cardiac repair, stem cell-based therapeutic strategies may become alternatives to donor heart transplantation. The principle of reprogramming adult terminally differentiated cells (iPSC) had a major impact on stem cell biology. One can now generate autologous pluripotent cells that highly resemble embryonic stem cells (ESC) and that are ethically inoffensive as opposed to human ESC. Yet, due to genetic and epigenetic aberrations arising during the full reprogramming process, it is questionable whether iPSC will enter the clinic in the near future. Therefore, the recent achievement of directly reprogramming fibroblasts into cardiomyocytes via a milder approach, thereby avoiding an initial pluripotent state, may become of great importance. In addition, various clinical scenarios will depend on the availability of specific cardiac cellular subtypes, for which a first step was achieved via our own programming approach to achieve cardiovascular cell subtypes. In this review, we discuss recent progress in the cardiovascular stem cell field addressing the above mentioned aspects.
Collapse
Affiliation(s)
- Robert David
- 1st Medical Department, University of Munich, Campus Grosshadern, Munich, Germany
| | | |
Collapse
|