1
|
Zhang Y, Lu Y, Hu X, Jiang M, Chen Z, Jin L, Li M, Chen C, Wang J. Functional characterization and therapeutic potential of human umbilical cord blood mononuclear cells. Regen Ther 2025; 28:101-114. [PMID: 40166041 PMCID: PMC11955793 DOI: 10.1016/j.reth.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 04/02/2025] Open
Abstract
Human umbilical cord blood mononuclear cells (hUCB-MNCs) are a population of cells derived from neonatal cord blood, encompassing various stem cells and immune cells. The unique characteristics of hUCB-MNCs endow them with distinctive multifunctionality, including the promotion of angiogenesis, acceleration of tissue repair, regulation of immune responses, neuroprotection, alleviation of inflammatory reactions, enhancement of antioxidant capacity, reduction of fibrosis processes, and inhibition of apoptosis. These diverse biological properties underscore the significant clinical therapeutic potential of hUCB-MNCs, which are widely applied in the treatment of various diseases. This review aims to summarize the underlying mechanisms responsible for the multifunctional attributes of hUCB-MNCs, elucidating their potential modes of action in disease management and providing novel theoretical insights and practical guidance for their expanded application across different disease domains. By synthesizing current research findings, this review may provide insights into the potential clinical applications of hUCB-MNCs in the fields of regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yueda Lu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xinyue Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Mengxing Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Zhixiu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Lingkun Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Min Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Chen Chen
- Department of Surgical Oncology, Anhui Provincial Children's Hospital, No.39 East Wangjiang Road, Hefei 230022, Anhui, China
| | - Jianye Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China
- The First Clinical College of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
- Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| |
Collapse
|
2
|
Makkaoui N, Prasad V, Bagchi P, Carmona T, Li K, Latham OL, Zhang Y, Lee J, Furdui CM, Maxwell JT. Cell-based therapies reverse the heart failure-altered right ventricular proteome towards a pre-disease state. Stem Cell Res Ther 2024; 15:420. [PMID: 39533351 PMCID: PMC11559167 DOI: 10.1186/s13287-024-04009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Congenital heart defects can lead to right ventricular (RV) pressure-overload and heart failure. Cell-based therapies, including mesenchymal stromal cells (MSCs) and c-kit positive cells (CPCs) have been studied clinically as options to restore heart function in disease states. Many studies have indicated these cells act through paracrine mechanisms to prevent apoptosis, promote cellular function, and regulate gene/protein expression. We aimed to determine the proteomic response of diseased hearts to cell therapy. METHODS We utilized a juvenile rat model of RV pressure overload created by banding the pulmonary artery (PAB). Two weeks post-banding, bone marrow-derived mesenchymal stromal cells (MSCs) and 3 populations of CPCs (nCPCs, cCPCs, ES-CPCs) were delivered to the RV free wall. RV function and cellular retention were measured for four weeks post-injection, at which point hearts were extracted and the RV was excised for liquid chromatography and tandem mass spectrometry. Resulting RV proteomes were compared and analyzed using systems biology and bioinformatics. RESULTS Proteomic profiling identified 1156 total proteins from the RV, of which 5.97% were significantly changed after PAB. This disease-altered proteome was responsive to cellular therapy, with 72% of the PAB-altered proteome being fully or partially reversed by MSC therapy. This was followed by nCPCs (54%), ES-CPCs (52%), and cCPCs (39%). Systems biology and bioinformatics analysis showed MSC, nCPC, or ES-CPC cell therapy is associated with a decrease in predicted adverse cardiac effects. We also observed an effect of cell therapy on the non-altered RV proteome, however, this was associated with minor predicted pathological endpoints. CONCLUSIONS Our data indicate MSCs, ES-CPCs, and nCPCs significantly reverse the PAB-altered proteome towards a pre-disease state in our animal model. These results indicate cell-based therapies show promise in improving RV function after pressure overload through partial restoration of the disease-altered cardiac proteome.
Collapse
Affiliation(s)
- Nour Makkaoui
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, 30322, USA
| | - Vidhya Prasad
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, 30322, USA
| | - Pritha Bagchi
- Emory Integrated Proteomics Core (EIPC), Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Tiffany Carmona
- Emory University College of Arts and Sciences, Atlanta, GA, 30322, USA
| | - Ke Li
- Emory University College of Arts and Sciences, Atlanta, GA, 30322, USA
| | - Olivia L Latham
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Jingyun Lee
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Joshua T Maxwell
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA.
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
3
|
Makkaoui N, Prasad V, Bagchi P, Carmona T, Li K, Latham O, Zhang Y, Lee J, Furdui C, Maxwell J. Cell-based therapies reverse the heart failure-altered right ventricular proteome. RESEARCH SQUARE 2024:rs.3.rs-4752035. [PMID: 39281857 PMCID: PMC11398576 DOI: 10.21203/rs.3.rs-4752035/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Congenital heart defects can lead to right ventricular (RV) pressure-overload and heart failure. Cell-based therapies, including mesenchymal stromal cells (MSCs) and c-kit positive cells (CPCs) have been studied clinically as options to restore heart function in disease states. Many studies have indicated these cells act through paracrine mechanisms to prevent apoptosis, promote cellular function, and regulate gene/protein expression. We aimed to determine the proteomic response of diseased hearts to cell therapy. Methods We utilized an animal model of RV pressure overload created by banding the pulmonary artery (PAB). Two weeks post-banding, bone marrow-derived mesenchymal stromal cells (MSCs) and 3 populations of CPCs (nCPCs, cCPCs, ES-CPCs) were delivered to the RV free wall. RV function and cellular retention were measured for four weeks post-injection, at which point hearts were extracted and the RV was excised for liquid chromatography and tandem mass spectrometry. Resulting RV proteomes were compared and analyzed using systems biology and bioinformatics. Results Proteomic profiling identified 1156 total proteins from the RV, of which 5.97% were significantly changed after PAB. This disease-altered proteome was responsive to cellular therapy, with 72% of the PAB-altered proteome being fully or partially reversed by MSC therapy. This was followed by nCPCs (54%), ES-CPCs (52%), and cCPCs (39%). Systems biology and bioinformatics analysis showed MSC, nCPC, or ES-CPC cell therapy is associated with a decrease in predicted adverse cardiac effects. We also observed an effect of cell therapy on the non-altered RV proteome, however, this was associated with minor predicted pathological endpoints. Conclusions Our data indicate MSCs, ES-CPCs, and nCPCs significantly reverse the PAB-altered proteome towards a pre-disease state. These results indicate cell-based therapies show promise in improving RV function after pressure overload through partial restoration of the disease-altered cardiac proteome.
Collapse
Affiliation(s)
| | | | | | | | - Ke Li
- Emory University Emory College of Arts and Sciences
| | | | | | | | | | | |
Collapse
|
4
|
Rubio-Ramos A, Labat-de-Hoz L, Correas I, Alonso MA. The MAL Protein, an Integral Component of Specialized Membranes, in Normal Cells and Cancer. Cells 2021; 10:1065. [PMID: 33946345 PMCID: PMC8145151 DOI: 10.3390/cells10051065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
The MAL gene encodes a 17-kDa protein containing four putative transmembrane segments whose expression is restricted to human T cells, polarized epithelial cells and myelin-forming cells. The MAL protein has two unusual biochemical features. First, it has lipid-like properties that qualify it as a member of the group of proteolipid proteins. Second, it partitions selectively into detergent-insoluble membranes, which are known to be enriched in condensed cell membranes, consistent with MAL being distributed in highly ordered membranes in the cell. Since its original description more than thirty years ago, a large body of evidence has accumulated supporting a role of MAL in specialized membranes in all the cell types in which it is expressed. Here, we review the structure, expression and biochemical characteristics of MAL, and discuss the association of MAL with raft membranes and the function of MAL in polarized epithelial cells, T lymphocytes, and myelin-forming cells. The evidence that MAL is a putative receptor of the epsilon toxin of Clostridium perfringens, the expression of MAL in lymphomas, the hypermethylation of the MAL gene and subsequent loss of MAL expression in carcinomas are also presented. We propose a model of MAL as the organizer of specialized condensed membranes to make them functional, discuss the role of MAL as a tumor suppressor in carcinomas, consider its potential use as a cancer biomarker, and summarize the directions for future research.
Collapse
Affiliation(s)
- Armando Rubio-Ramos
- Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.R.-R.); (L.L.-d.-H.); (I.C.)
| | - Leticia Labat-de-Hoz
- Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.R.-R.); (L.L.-d.-H.); (I.C.)
| | - Isabel Correas
- Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.R.-R.); (L.L.-d.-H.); (I.C.)
- Department of Molecular Biology, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Miguel A. Alonso
- Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.R.-R.); (L.L.-d.-H.); (I.C.)
| |
Collapse
|
5
|
Zhao L, Cheng G, Choksi K, Samanta A, Girgis M, Soder R, Vincent RJ, Wulser M, De Ruyter M, McEnulty P, Hauptman J, Yang Y, Weiner CP, Dawn B. Transplantation of Human Umbilical Cord Blood-Derived Cellular Fraction Improves Left Ventricular Function and Remodeling After Myocardial Ischemia/Reperfusion. Circ Res 2019; 125:759-772. [PMID: 31462157 DOI: 10.1161/circresaha.119.315216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Rationale: Human umbilical cord blood (hUCB) contains diverse populations of stem/progenitor cells. Whether hUCB-derived nonhematopoietic cells would induce cardiac repair remains unknown. Objective: To examine whether intramyocardial transplantation of hUCB-derived CD45-Lin- nonhematopoietic cellular fraction after a reperfused myocardial infarction in nonimmunosuppressed rats would improve cardiac function and ameliorate ventricular remodeling. Methods and Results: Nonhematopoietic CD45-Lin- cells were isolated from hUCB. Flow cytometry and quantitative polymerase chain reaction were used to characterize this subpopulation. Age-matched male Fischer 344 rats underwent a 30-minute coronary occlusion followed by reperfusion and 48 hours later received intramyocardial injection of vehicle or hUCB CD45-Lin- cells. After 35 days, compared with vehicle-treated rats, CD45-Lin- cell-treated rats exhibited improved left ventricular function, blunted left ventricular hypertrophy, greater preservation of viable myocardium in the infarct zone, and superior left ventricular remodeling. Mechanistically, hUCB CD45-Lin- cell injection favorably modulated molecular pathways regulating myocardial fibrosis, cardiomyocyte apoptosis, angiogenesis, and inflammation in postinfarct ventricular myocardium. Rare persistent transplanted human cells could be detected at both 4 and 35 days after myocardial infarction. Conclusions: Transplantation of hUCB-derived CD45-Lin- nonhematopoietic cellular subfraction after a reperfused myocardial infarction in nonimmunosuppressed rats ameliorates left ventricular dysfunction and improves remodeling via favorable paracrine modulation of molecular pathways. These findings with human cells in a clinically relevant model of myocardial ischemia/reperfusion in immunocompetent animals may have significant translational implications.Visual Overview: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Lin Zhao
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Guangming Cheng
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Kashyap Choksi
- Cardiology Consultants of South Georgia, Thomasville (K.C.)
| | - Anweshan Samanta
- Department of Internal Medicine (A.S.), University of Missouri-Kansas City
| | - Magdy Girgis
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Rupal Soder
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Robert J Vincent
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Michael Wulser
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Matt De Ruyter
- Department of Orthopedic Surgery (M.D.R.), University of Missouri-Kansas City
| | - Patrick McEnulty
- Department of Radiology, University of Kansas School of Medicine-Wichita (P.M.)
| | - Jeryl Hauptman
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Yanjuan Yang
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| | - Carl P Weiner
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City (R.S., R.J.V., M.W., C.P.W.)
| | - Buddhadeb Dawn
- From the Department of Internal Medicine, University of Nevada, Las Vegas School of Medicine (L.Z., G.C., M.G., J.H., Y.Y., B.D.)
| |
Collapse
|
6
|
Burkhart HM, Qureshi MY, Rossano JW, Cantero Peral S, O'Leary PW, Hathcock M, Kremers W, Nelson TJ. Autologous stem cell therapy for hypoplastic left heart syndrome: Safety and feasibility of intraoperative intramyocardial injections. J Thorac Cardiovasc Surg 2019; 158:1614-1623. [PMID: 31345560 DOI: 10.1016/j.jtcvs.2019.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Staged surgical palliation for hypoplastic left heart syndrome results in an increased workload on the right ventricle serving as the systemic ventricle. Concerns for cardiac dysfunction and long-term heart failure have generated interest in first-in-infant, cell-based therapies as an additional surgical treatment modality. METHODS A phase 1 clinical trial was conducted to evaluate the safety and feasibility of direct intramyocardial injection of autologous umbilical cord blood-derived mononuclear cells in 10 infants with hypoplastic left heart syndrome at the time of stage II palliation. RESULTS All 10 patients underwent successful stage II palliation and intramyocardial injection of umbilical cord blood-derived mononuclear cells. Operative mortality was 0%. There was a single adverse event related to cell delivery: An injection site epicardial bleed that required simple oversew. The cohort did not demonstrate any significant safety concerns over 6 months. Additionally, the treatment group did not demonstrate any reduction in cardiac function in the context of the study related intramyocardial injections of autologous cells. CONCLUSIONS This phase 1 clinical trial showed that delivering autologous umbilical cord blood-derived mononuclear cells directly into the right ventricular myocardium during planned stage II surgical palliation for hypoplastic left heart syndrome was safe and feasible. Secondary findings of preservation of baseline right ventricular function throughout follow-up and normalized growth rates support the design of a phase 2b follow-up trial.
Collapse
Affiliation(s)
- Harold M Burkhart
- Division of Cardiovascular and Thoracic Surgery, University of Oklahoma, Oklahoma City, Okla.
| | | | - Joseph W Rossano
- Cardiac Center, Children's Hospital of Philadelphia, Philadelphia, Pa
| | | | | | - Matthew Hathcock
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minn
| | - Walter Kremers
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minn
| | - Timothy J Nelson
- Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minn; Division of General Internal Medicine, Mayo Clinic, Rochester, Minn; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minn; Center for Regenerative Medicine, Mayo Clinic, Rochester, Minn
| |
Collapse
|
7
|
Abstract
This article presents the case of a newly born female patient with a cleft of the primary palate (alveolar cleft), with an in utero diagnosis of the described cleft, from whom umbilical cord stem cells are obtained and cryopreserved. The patient is managed with nasoalveolar molding, and at 5 months of age, she is taken to surgery for cheiloplasty and gingivoperiostioplasty with umbilical cord stem cells. A radiographic and CT follow-up is carried out on the described cleft.
Collapse
|
8
|
Expanded CD133 + Cells from Human Umbilical Cord Blood Improved Heart Function in Rats after Severe Myocardial Infarction. Stem Cells Int 2018; 2018:5412478. [PMID: 29760727 PMCID: PMC5925035 DOI: 10.1155/2018/5412478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/08/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Pharmacological approaches are partially effective in limiting infarct size. Cell therapies using a cell population enriched with endothelial progenitor cells (EPCs) CD133+ have opened new perspectives for the treatment of ischemic areas after infarction. This preclinical study evaluated the effect of intramyocardial transplantation of purified or expanded human umbilical cord blood-derived CD133+ cells on the recovery of rats following acute myocardial infarction (AMI). Histology studies, electrocardiogram, and fluorescence in situ hybridization (FISH) were used to evaluate heart recovery. Purified CD133+ cells, enriched in endothelial progenitor cells, when expanded in vitro acquired an endothelial-like cell phenotype expressing CD31 and von Willebrand factor (vWF). The group of infarcted rats that received expanded CD133+ cells had a more significant recovery of contraction performance and less heart remodeling than the group that received purified CD133+ cells. Either purified or expanded CD133+ cells were able to induce neovascularization in the infarcted myocardium in an equivalent manner. Few human cells were detected in the infarcted myocardium of the rats 28 days after transplantation suggesting that the effects observed might be related primarily to paracrine activity. Although both cell populations ameliorated the infarcted heart and are suitable for regeneration of the vascular system, expanded CD133+ cells are more beneficial and promising candidates for vascular regeneration.
Collapse
|
9
|
Shafei AES, Ali MA, Ghanem HG, Shehata AI, Abdelgawad AA, Handal HR, ElSayed AS, Ashaal AE, Ali MM, El-Shal AS. Mechanistic effects of mesenchymal and hematopoietic stem cells: New therapeutic targets in myocardial infarction. J Cell Biochem 2018; 119:5274-5286. [PMID: 29266431 DOI: 10.1002/jcb.26637] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022]
Abstract
Myocardial infarction (MI) results in dysfunction and irreversible loss of cardiomyocytes and is of the most serious health threats today. Mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) have been explored as promising cell therapy in MI and regenerative therapy. Recently, reports investigated the potential therapeutic effects of MSCs or HSCs transplantation after MI in numerous experimental and clinical studies; however, their results are controversy and needs more explorations. The current review is an attempt to clarify the therapeutic potentials of MSCs and HSCs in MI therapy, as well as their possible effects; especially the paracrine one and the exosome-derived stem cell among animal models as well as clinical trials conducted within the last 10 years. In this context, various sources of MSCs and HSCs have been addressed in helping cardiac regeneration by either revitalizing the cardiac stem cells niche or revascularizing the arteries and veins of the heart. In addition, both MSCs and HSCs could produce paracrine mediators and growth factors which led to cardiomyocytes protection, angiogenesis, immunemodulation, antioxidants, anti-apoptotic, anti-inflammatory, antifibrotic, as well as increasing cardiac contractility. Recently, microRNAs (miRNAs), post-transcriptional regulators of gene expression, and long non-coding RNA (lncRNA), a miRNA sponge, are recent stem cell-derived mediators can be promising targets of MSCs and HSCs through their paracrine effects. Although MSCs and HSCs have achieved considerable achievements, however, some challenges still remain that need to be overcome in order to establish it as a successful technique. The present review clarified the mechanistic potentials of MSCs and HSCs especially paracrine effects involved in MI including human and animal studies and the challenges challenges regarding type, differentiation, route, and number of injections.
Collapse
Affiliation(s)
- Ayman El-Sayed Shafei
- Biomedical Research Department, Military Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mahmoud A Ali
- Biomedical Research Department, Military Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Hazem G Ghanem
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | - Ahmed I Shehata
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | | | - Hossam R Handal
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | | | - Ahmed E Ashaal
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | - Mazen M Ali
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | - Amal S El-Shal
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
Shafei AES, Ali MA, Ghanem HG, Shehata AI, Abdelgawad AA, Handal HR, Talaat KA, Ashaal AE, El-Shal AS. Mesenchymal stem cell therapy: A promising cell-based therapy for treatment of myocardial infarction. J Gene Med 2017; 19. [PMID: 29044850 DOI: 10.1002/jgm.2995] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/07/2017] [Accepted: 10/07/2017] [Indexed: 12/12/2022] Open
Abstract
For decades, mesenchymal stem (MSCs) cells have been used for cardiovascular diseases as regenerative therapy. This review is an attempt to summarize the types of MSCs involved in myocardial infarction (MI) therapy, as well as its possible mechanisms effects, especially the paracrine one in MI focusing on the studies (human and animal) conducted within the last 10 years. Recently, reports showed that MSC therapy could have infarct-limiting effects after MI in both experimental and clinical trials. In this context, various types of MSCs can help cardiac regeneration by either revitalizing the cardiac stem cells or revascularizing the arteries and veins of the heart. Furthermore, MSCs could produce paracrine growth factors that increase the survival of nearby cardiomyocytes, as well as increase angiogenesis through recruitment of stem cell from bone marrow or inducing vessel growth from existing capillaries. Recent research suggests that the paracrine effects of MSCs could be mediated by extracellular vesicles including exosomes. Exosomal microRNAs (miRNAs) released by MSCs are promising therapeutic hotspot target for MI. This could be attributed to the role of miRNA in cardiac biology, including cardiac regeneration, stem cell differentiation, apoptosis, neovascularization, cardiac contractility and cardiac remodeling. Furthermore, gene-modified MSCs could be a recent promising therapy for MI to enhance the paracrine effects of MSCs, including better homing and effective cell targeted tissue regeneration. Although MSC therapy has achieved considerable attention and progress, there are critical challenges that remains to be overcome to achieve the most effective successful cell-based therapy in MI.
Collapse
Affiliation(s)
- Ayman El-Sayed Shafei
- Biomedical Research Department, Military Armed Forces College of Medicine, Cairo, Egypt
| | - Mahmoud Ahmed Ali
- Biomedical Research Department, Military Armed Forces College of Medicine, Cairo, Egypt
| | | | | | | | | | | | | | - Amal S El-Shal
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
11
|
Abstract
For >4 decades, the holy grail in the treatment of acute myocardial infarction has been the mitigation of lethal injury. Despite promising initial results and decades of investigation by the cardiology research community, the only treatment with proven efficacy is early reperfusion of the occluded coronary artery. The remarkable record of failure has led us and others to wonder if cardioprotection is dead. The path to translation, like the ascent to Everest, is certainly littered with corpses. We do, however, highlight a therapeutic principle that provides a glimmer of hope: cellular postconditioning. Administration of cardiosphere-derived cells after reperfusion limits infarct size measured acutely, while providing long-term structural and functional benefits. The recognition that cell therapy may be cardioprotective, and not just regenerative, merits further exploration before we abandon the pursuit entirely.
Collapse
Affiliation(s)
- David J Lefer
- From Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans (D.J.L.); and Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.).
| | - Eduardo Marbán
- From Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans (D.J.L.); and Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.)
| |
Collapse
|
12
|
Newcomb JD, Sanberg PR, Klasko SK, Willing AE. Umbilical Cord Blood Research: Current and Future Perspectives. Cell Transplant 2017. [DOI: 10.3727/000000007783464623] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Umbilical cord blood (UCB) banking has become a new obstetrical trend. It offers expectant parents a biological insurance policy that can be used in the event of a child or family member's life-threatening illness and puts patients in a position of control over their own treatment options. However, its graduation to conventional therapy in the clinical realm relies on breakthrough research that will prove its efficacy for a range of ailments. Expanding the multipotent cells found within the mononuclear fraction of UCB so that adequate dosing can be achieved, effectively expanding desired cells ex vivo, establishing its safety and limitations in HLA-mismatched recipients, defining its mechanisms of action, and proving its utility in a wide variety of both rare and common illnesses and diseases are a few of the challenges left to tackle. Nevertheless, the field is moving fast and new UCB-based therapies are on the horizon.
Collapse
Affiliation(s)
- Jennifer D. Newcomb
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, College of Medicine, Tampa, FL 33612, USA
| | - Paul R. Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, College of Medicine, Tampa, FL 33612, USA
| | - Stephen K. Klasko
- Department of Obstetrics and Gynecology, University of South Florida, College of Medicine, Tampa, FL 33612, USA
| | - Alison E. Willing
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
13
|
Borlongan CV, Yu G, Matsukawa N, Yasuhara T, Hara K, Xu L. Article Commentary: Cell Transplantation: Stem Cells in the Spotlight. Cell Transplant 2017; 14:519-526. [DOI: 10.3727/000000005783982774] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Cesar V. Borlongan
- Neurology/Insttitute of Molecular Medicind & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA, USA
- Research/Affiliations Service Line, Augusta VAMC, Augusta, GA, USA
| | - Guolong Yu
- Neurology/Insttitute of Molecular Medicind & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA, USA
- Research/Affiliations Service Line, Augusta VAMC, Augusta, GA, USA
| | - Noriyuki Matsukawa
- Neurology/Insttitute of Molecular Medicind & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA, USA
- Research/Affiliations Service Line, Augusta VAMC, Augusta, GA, USA
| | - Takao Yasuhara
- Neurology/Insttitute of Molecular Medicind & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA, USA
- Research/Affiliations Service Line, Augusta VAMC, Augusta, GA, USA
| | - Koichi Hara
- Neurology/Insttitute of Molecular Medicind & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA, USA
- Research/Affiliations Service Line, Augusta VAMC, Augusta, GA, USA
| | - Lin Xu
- Neurology/Insttitute of Molecular Medicind & Genetics/School of Graduate Studies, Medical College of Georgia, Augusta, GA, USA
- Research/Affiliations Service Line, Augusta VAMC, Augusta, GA, USA
| |
Collapse
|
14
|
SanMartin A, Borlongan CV. Article Commentary: Cell Transplantation: Toward Cell Therapy. Cell Transplant 2017; 15:665-73. [PMID: 17176618 DOI: 10.3727/000000006783981666] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Agneta SanMartin
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL 33612, USA.
| | | |
Collapse
|
15
|
Newcomb JD, Ajmo CT, Sanberg CD, Sanberg PR, Pennypacker KR, Willing AE. Timing of Cord Blood Treatment after Experimental Stroke Determines Therapeutic Efficacy. Cell Transplant 2017; 15:213-23. [PMID: 16719056 DOI: 10.3727/000000006783982043] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Embolic stroke is thought to cause irreparable damage in the brain immediately adjacent to the region of reduced blood perfusion. Therefore, much of the current research focuses on treatments such as anti-inflammatory, neuroprotective, and cell replacement strategies to minimize behavioral and physiological consequences. In the present study, intravenous delivery of human umbilical cord blood cells (HUCBC) 48 h after a middle cerebral artery occlusion (MCAo) in a rat resulted in both behavioral and physiological recovery. Nissl and TUNEL staining demonstrated that many of the neurons in the core were rescued, indicating that while both necrotic and apoptotic cell death occur in ischemia, it is clear that apoptosis plays a larger role than first anticipated. Further, immunohistochemical and histochemical analysis showed a diminished and/or lack of granulocyte and monocyte infiltration and astrocytic and microglial activation in the parenchyma in animals treated with HUCBC 48 h poststroke. Successful treatment at this time point should offer encouragement to clinicians that a therapy with a broader window of efficacy may soon be available to treat stroke.
Collapse
Affiliation(s)
- Jennifer D Newcomb
- Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | | | | | | | | | |
Collapse
|
16
|
Yousefi B, Sanooghi D, Faghihi F, Joghataei MT, Latifi N. Evaluation of motor neuron differentiation potential of human umbilical cord blood- derived mesenchymal stem cells, in vitro. J Chem Neuroanat 2017; 81:18-26. [PMID: 28153469 DOI: 10.1016/j.jchemneu.2017.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/25/2016] [Accepted: 01/22/2017] [Indexed: 10/20/2022]
Abstract
Many people suffer from spinal cord injuries annually. These deficits usually threaten the quality of life of patients. As a postpartum medically waste product, human Umbilical Cord Blood (UCB) is a rich source of stem cells with self- renewal properties and neural differentiation capacity which made it useful in regenerative medicine. Since there is no report on potential of human umbilical cord blood-derived mesenchymal stem cells into motor neurons, we set out to evaluate the differentiation properties of these cells into motor neuron-like cells through administration of Retinoic Acid(RA), Sonic Hedgehog(Shh) and BDNF using a three- step in vitro procedure. The results were evaluated using Real-time PCR, Flowcytometry and Immunocytochemistry for two weeks. Our data showed that the cells changed into bipolar morphology and could express markers related to motor neuron; including Hb-9, Pax-6, Islet-1, NF-H, ChAT at the level of mRNA and protein. We could also quantitatively evaluate the expression of Islet-1, ChAT and NF-H at 7 and 14days post- induction using flowcytometry. It is concluded that human UCB-MSCs is potent to express motor neuron- related markers in the presence of RA, Shh and BDNF through a three- step protocol; thus it could be a suitable cell candidate for regeneration of motor neurons in spinal cord injuries.
Collapse
Affiliation(s)
- Behnam Yousefi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Sanooghi
- Department of Genetics, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Nourahmad Latifi
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Zhou N, Wang J, Li X, Zhao Y, Sun Y, Zou C. Hetrombopag, a Thrombopoietin Receptor Agonist, Protects Cardiomyocyte Survival from Oxidative Stress Damage as an Enhancer of Stem Cells. Cardiovasc Drugs Ther 2016; 30:567-577. [PMID: 27838864 DOI: 10.1007/s10557-016-6696-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE Current human umbilical cord blood stem cell therapy faces the great challenges, because the stem cells are scarce and cannot survive for a long time. Here we describe how hetrombopag, an orally-active TPO receptor agonists, enhanced ex vivo expansion of human UCB stem cells, and protected cardiac myocytes from the damage caused by oxidative stress. METHODS Ex vivo expansion of stem cells were performed in serum-free medium supplemented with rhSCF and rhFL plus hetrombopag for 7 days. The percentage and number of stem cell subsets were determined by flow cytometry. Rat cardiac myocytes, ex vivo expanded stem cells, or cardiac myocytes plus ex vivo expanded stem cells were serum starved for 24 hours, and were then subjected to H2O2, hetrombopag or both for 12 hours at the indicated concentrations. Cell viability assays, protein microarrays and western blots were then performed in each group. RESULTS Our studies first revealed that the combination of hetrombopag and rhTPO manifested additive effect on ex vivo expansion of human UCB stem cells. Besides, hetrombopag dose-dependently enhanced the beneficial effects of ex vivo expanded human UCB MNCs in increasing the survival of injured cardiomyocytes during free oxygen radical stress. CONCLUSION These data, for the first time, uncovered a novel function of non-peptide small molecular TPO receptor agonists as enhancers of stem cells in protecting cardiac myocyte survival from oxidative stress damage, which might provide a new therapeutic avenue for the treatment of oxidative stress-related cardiovascular disease. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Nannan Zhou
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Jianchun Wang
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Xiaodong Li
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Yong Zhao
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Yuanyuan Sun
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China
| | - Chengwei Zou
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
18
|
Henning RJ. Therapeutic angiogenesis: angiogenic growth factors for ischemic heart disease. Future Cardiol 2016; 12:585-99. [PMID: 27420190 DOI: 10.2217/fca-2016-0006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stem cells encode vascular endothelial growth factors (VEGFs), fibroblastic growth factors (FGFs), stem cell factor, stromal cell-derived factor, platelet growth factor and angiopoietin that can contribute to myocardial vascularization. VEGFs and FGFs are the most investigated growth factors. VEGFs regulate angiogenesis and vasculogenesis. FGFs stimulate vessel cell proliferation and differentiation and are regulators of endothelial cell migration, proliferation and survival. Clinical trials of VEGF or FGF for myocardial angiogenesis have produced disparate results. The efficacy of therapeutic angiogenesis can be improved by: (1) identifying the most optimal patients; (2) increased knowledge of angiogenic factor pharmacokinetics and proper dose; (3) prolonging contact of angiogenic factors with the myocardium; (4) increasing the efficiency of VEGF or FGF gene transduction; and (5) utilizing PET or MRI to measure myocardial perfusion and perfusion reserve.
Collapse
Affiliation(s)
- Robert J Henning
- The University of South Florida and the James A. Haley Hospital, Tampa, FL 33612 USA
| |
Collapse
|
19
|
Stem Cell Banking and Its Impact on Cardiac Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 951:163-178. [PMID: 27837563 DOI: 10.1007/978-3-319-45457-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cardiovascular diseases, including heart failure, are the most frequent cause of death annually, even higher than any other pathologies. Specifically, patients who suffer from myocardial infarction may encounter adverse remodeling processes of the heart that can ultimately lead to heart failure. Prognosis of patients affected by heart failure is very poor with 5-year mortality close to 50 %. Despite the impressive progress in the clinical treatment of heart failure in recent years, heart transplantation is still required to avoid death as the result of the inexorable decline in cardiac function. Unfortunately, the availability of donor human hearts for transplantation largely fails to cover the number of potential recipient requests. From this urgent unmet clinical need the interest in stem cell applications for heart regeneration made its start, and has rapidly grown in the last decades. Indeed, the discovery and application of stem and progenitor cells as therapeutic agents has raised substantial interest with the objective of reversing these processes, and ultimately inducing cardiac regeneration. In this scenario, the role of biobanking may play a remarkable role to provide cells at the right time according to the patient's clinical needs, mostly for autologous use in the acute setting of myocardial infarction, largely reducing the time needed for cell preparation and expansion before administration.
Collapse
|
20
|
Le-Buu Pham T, Nguyen TT, Thi-Van Bui A, Pham HT, Phan NK, Thi-Thu Nguyen M, Van Pham P. Preliminary evaluation of treatment efficacy of umbilical cord blood-derived mesenchymal stem cell-differentiated cardiac progenitor cells in a myocardial injury mouse model. BIOMEDICAL RESEARCH AND THERAPY 2015. [DOI: 10.7603/s40730-015-0032-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
21
|
Laiva AL, Venugopal JR, Navaneethan B, Karuppuswamy P, Ramakrishna S. Biomimetic approaches for cell implantation to the restoration of infarcted myocardium. Nanomedicine (Lond) 2015; 10:2907-30. [PMID: 26371367 DOI: 10.2217/nnm.15.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Compelling evidences accumulated over the years have proven stem cells as a promising source for regenerative medicine. However, the inadequacy with the design of delivery modalities has prolonged the research in realizing an ideal cell-based approach for the regeneration of infarcted myocardium. Currently, some modest improvements in cardiac function have been documented in clinical trials with stem cell treatments, although regenerating a fully functional myocardium remains a dream for cardiac surgeons. This review provides an overview on the significance of stem cell therapy, the current attempts to resolve the drawbacks with the cell implantation approach and the various stratagems adopted with electrospun hybrid nanofibers for implementation in myocardial regenerative therapy.
Collapse
Affiliation(s)
- Ashang Luwang Laiva
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576.,Amity Institute of Nanotechnology, Amity University, Noida, UP, India
| | - Jayarama Reddy Venugopal
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| | - Balchandar Navaneethan
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| | - Priyadharsini Karuppuswamy
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| | - Seeram Ramakrishna
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| |
Collapse
|
22
|
Porada CD, Atala AJ, Almeida-Porada G. The hematopoietic system in the context of regenerative medicine. Methods 2015; 99:44-61. [PMID: 26319943 DOI: 10.1016/j.ymeth.2015.08.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/06/2015] [Accepted: 08/23/2015] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cells (HSC) represent the prototype stem cell within the body. Since their discovery, HSC have been the focus of intensive research, and have proven invaluable clinically to restore hematopoiesis following inadvertent radiation exposure and following radio/chemotherapy to eliminate hematologic tumors. While they were originally discovered in the bone marrow, HSC can also be isolated from umbilical cord blood and can be "mobilized" peripheral blood, making them readily available in relatively large quantities. While their ability to repopulate the entire hematopoietic system would already guarantee HSC a valuable place in regenerative medicine, the finding that hematopoietic chimerism can induce immunological tolerance to solid organs and correct autoimmune diseases has dramatically broadened their clinical utility. The demonstration that these cells, through a variety of mechanisms, can also promote repair/regeneration of non-hematopoietic tissues as diverse as liver, heart, and brain has further increased their clinical value. The goal of this review is to provide the reader with a brief glimpse into the remarkable potential HSC possess, and to highlight their tremendous value as therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| | - Anthony J Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, 391 Technology Way, Winston-Salem, NC 27157-1083, United States.
| |
Collapse
|
23
|
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
|
24
|
Suss PH, Capriglione LGA, Barchiki F, Miyague L, Jackowski D, Fracaro L, Schittini AV, Senegaglia AC, Rebelatto CLK, Olandoski M, Correa A, Brofman PRS. Direct intracardiac injection of umbilical cord-derived stromal cells and umbilical cord blood-derived endothelial cells for the treatment of ischemic cardiomyopathy. Exp Biol Med (Maywood) 2015; 240:969-78. [PMID: 25576340 DOI: 10.1177/1535370214565077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/12/2014] [Indexed: 01/09/2023] Open
Abstract
The development of new therapeutic strategies is necessary to reduce the worldwide social and economic impact of cardiovascular disease, which produces high rates of morbidity and mortality. A therapeutic option that has emerged in the last decade is cell therapy. The aim of this study was to compare the effect of transplanting human umbilical cord-derived stromal cells (UCSCs), human umbilical cord blood-derived endothelial cells (UCBECs) or a combination of these two cell types for the treatment of ischemic cardiomyopathy (IC) in a Wistar rat model. IC was induced by left coronary artery ligation, and baseline echocardiography was performed seven days later. Animals with a left ventricular ejection fraction (LVEF) of ≤40% were selected for the study. On the ninth day after IC was induced, the animals were randomized into the following experimental groups: UCSCs, UCBECs, UCSCs plus UCBECs, or vehicle (control). Thirty days after treatment, an echocardiographic analysis was performed, followed by euthanasia. The animals in all of the cell therapy groups, regardless of the cell type transplanted, had less collagen deposition in their heart tissue and demonstrated a significant improvement in myocardial function after IC. Furthermore, there was a trend of increasing numbers of blood vessels in the infarcted area. The median value of LVEF increased by 7.19% to 11.77%, whereas the control group decreased by 0.24%. These results suggest that UCSCs and UCBECs are promising cells for cellular cardiomyoplasty and can be an effective therapy for improving cardiac function following IC.
Collapse
Affiliation(s)
- Paula H Suss
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | | | - Fabiane Barchiki
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Lye Miyague
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Danielle Jackowski
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Letícia Fracaro
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Andressa V Schittini
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ/PR, Curitiba 81.350-010, Brazil
| | - Alexandra C Senegaglia
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Carmen L K Rebelatto
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Márcia Olandoski
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| | - Alejandro Correa
- Carlos Chagas Institute, Oswaldo Cruz Foundation, FIOCRUZ/PR, Curitiba 81.350-010, Brazil
| | - Paulo R S Brofman
- Core for Cell Technology, Pontifícia Universidade Católica do Paraná, Curitiba 80.215-901, Brazil
| |
Collapse
|
25
|
Cantero Peral S, Burkhart HM, Oommen S, Yamada S, Nyberg SL, Li X, O'Leary PW, Terzic A, Cannon BC, Nelson TJ. Safety and feasibility for pediatric cardiac regeneration using epicardial delivery of autologous umbilical cord blood-derived mononuclear cells established in a porcine model system. Stem Cells Transl Med 2015; 4:195-206. [PMID: 25561683 DOI: 10.5966/sctm.2014-0195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Congenital heart diseases (CHDs) requiring surgical palliation mandate new treatment strategies to optimize long-term outcomes. Despite the mounting evidence of cardiac regeneration, there are no long-term safety studies of autologous cell-based transplantation in the pediatric setting. We aimed to establish a porcine pipeline to evaluate the feasibility and long-term safety of autologous umbilical cord blood mononuclear cells (UCB-MNCs) transplanted into the right ventricle (RV) of juvenile porcine hearts. Piglets were born by caesarean section to enable UCB collection. Upon meeting release criteria, 12 animals were randomized in a double-blinded fashion prior to surgical delivery of test article (n=6) or placebo (n=6). The UCB-MNC (3×10(6) cells per kilogram) or control (dimethyl sulfoxide, 10%) products were injected intramyocardially into the RV under direct visualization. The cohorts were monitored for 3 months after product delivery with assessments of cardiac performance, rhythm, and serial cardiac biochemical markers, followed by terminal necropsy. No mortalities were associated with intramyocardial delivery of UCB-MNCs or placebo. Two animals from the placebo group developed local skin infection after surgery that responded to antibiotic treatment. Electrophysiological assessments revealed no arrhythmias in either group throughout the 3-month study. Two animals in the cell-therapy group had transient, subclinical dysrhythmia in the perioperative period, likely because of an exaggerated response to anesthesia. Overall, this study demonstrated that autologous UCB-MNCs can be safely collected and surgically delivered in a pediatric setting. The safety profile establishes the foundation for cell-based therapy directed at the RV of juvenile hearts and aims to accelerate cell-based therapies toward clinical trials for CHD.
Collapse
Affiliation(s)
- Susana Cantero Peral
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Harold M Burkhart
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Saji Oommen
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Satsuki Yamada
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Scott L Nyberg
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Xing Li
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Patrick W O'Leary
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Andre Terzic
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Bryan C Cannon
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Timothy J Nelson
- Division of General Internal Medicine, Center for Regenerative Medicine, Pediatric Cardiothoracic Surgery, Division of Cardiovascular Diseases, Transplant Center, Division of Biomedical Statistics and Informatics, Division of Pediatric Cardiology, Department of Molecular Pharmacology and Experimental Therapeutics, and Mayo Clinic, Rochester, Minnesota, USA; Program of Doctorate of Internal Medicine, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
26
|
Henning RJ, Sanberg P, Jimenez E. Human cord blood stem cell paracrine factors activate the survival protein kinase Akt and inhibit death protein kinases JNK and p38 in injured cardiomyocytes. Cytotherapy 2014; 16:1158-68. [DOI: 10.1016/j.jcyt.2014.01.415] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/06/2014] [Accepted: 01/30/2014] [Indexed: 01/08/2023]
|
27
|
Chen J, Venkat P, Zacharek A, Chopp M. Neurorestorative therapy for stroke. Front Hum Neurosci 2014; 8:382. [PMID: 25018718 PMCID: PMC4072966 DOI: 10.3389/fnhum.2014.00382] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 05/14/2014] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is responsible for many deaths and long-term disability world wide. Development of effective therapy has been the target of intense research. Accumulating preclinical literature has shown that substantial functional improvement after stroke can be achieved using subacutely administered cell-based and pharmacological therapies. This review will discuss some of the latest findings on bone marrow-derived mesenchymal stem cells (BMSCs), human umbilical cord blood cells, and off-label use of some pharmacological agents, to promote recovery processes in the sub-acute and chronic phases following stroke. This review paper also focuses on molecular mechanisms underlying the cell-based and pharmacological restorative processes, which enhance angiogenesis, arteriogenesis, neurogenesis, and white matter remodeling following cerebral ischemia as well as an analysis of the interaction/coupling among these restorative events. In addition, the role of microRNAs mediating the intercellular communication between exogenously administered cells and parenchymal cells, and their effects on the regulation of angiogenesis and neuronal progenitor cell proliferation and differentiation, and brain plasticity after stroke are described.
Collapse
Affiliation(s)
- Jieli Chen
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA ; Department of Physics, Oakland University , Rochester, MI , USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA ; Department of Physics, Oakland University , Rochester, MI , USA
| |
Collapse
|
28
|
Stem Cell Banking for Regenerative and Personalized Medicine. Biomedicines 2014; 2:50-79. [PMID: 28548060 PMCID: PMC5423479 DOI: 10.3390/biomedicines2010050] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/10/2014] [Accepted: 02/17/2014] [Indexed: 12/12/2022] Open
Abstract
Regenerative medicine, tissue engineering and gene therapy offer the opportunity to treat and cure many of today’s intractable afflictions. These approaches to personalized medicine often utilize stem cells to accomplish these goals. However, stem cells can be negatively affected by donor variables such as age and health status at the time of collection, compromising their efficacy. Stem cell banking offers the opportunity to cryogenically preserve stem cells at their most potent state for later use in these applications. Practical stem cell sources include bone marrow, umbilical cord blood and tissue, and adipose tissue. Each of these sources contains stem cells that can be obtained from most individuals, without too much difficulty and in an economical fashion. This review will discuss the advantages and disadvantages of each stem cell source, factors to be considered when contemplating banking each stem cell source, the methodology required to bank each stem cell source, and finally, current and future clinical uses of each stem cell source.
Collapse
|
29
|
Biazar E. Use of umbilical cord and cord blood-derived stem cells for tissue repair and regeneration. Expert Opin Biol Ther 2014; 14:301-10. [PMID: 24456082 DOI: 10.1517/14712598.2014.867943] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Potential use of umbilical cord (UC) is one of the most exciting frontiers in medicine for repairing damaged tissues. UC and cord blood-derived stem cells are the world's largest potential sources of stem cells. UC contains a mixture of stem and progenitor cells at different lineage commitment stages and UC has been verified as a candidate for cell-based therapies and tissue engineering applications due to the capability of these cells for extensive self-renewal and multi-lineage character in differentiation potential. AREAS COVERED UC-based repair or regeneration of organs (i.e., heart, nerve, skin, etc.) is a high-priority research worldwide. EXPERT OPINION The aim of this review is to summarize the knowledge about UC with main focus on its applications for tissue repair and regeneration.
Collapse
Affiliation(s)
- Esmaeil Biazar
- Islamic Azad University, Department of Biomedical Engineering, Tonekabon Branch , Tonekabon , Iran +00981924271105 ;
| |
Collapse
|
30
|
Liu X, Ye R, Yan T, Yu SP, Wei L, Xu G, Fan X, Jiang Y, Stetler RA, Liu G, Chen J. Cell based therapies for ischemic stroke: from basic science to bedside. Prog Neurobiol 2013; 115:92-115. [PMID: 24333397 DOI: 10.1016/j.pneurobio.2013.11.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022]
Abstract
Cell therapy is emerging as a viable therapy to restore neurological function after stroke. Many types of stem/progenitor cells from different sources have been explored for their feasibility and efficacy for the treatment of stroke. Transplanted cells not only have the potential to replace the lost circuitry, but also produce growth and trophic factors, or stimulate the release of such factors from host brain cells, thereby enhancing endogenous brain repair processes. Although stem/progenitor cells have shown a promising role in ischemic stroke in experimental studies as well as initial clinical pilot studies, cellular therapy is still at an early stage in humans. Many critical issues need to be addressed including the therapeutic time window, cell type selection, delivery route, and in vivo monitoring of their migration pattern. This review attempts to provide a comprehensive synopsis of preclinical evidence and clinical experience of various donor cell types, their restorative mechanisms, delivery routes, imaging strategies, future prospects and challenges for translating cell therapies as a neurorestorative regimen in clinical applications.
Collapse
Affiliation(s)
- Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Neurology, Tianjin General Hospital, Tianjin University School of Medicine, Tianjin, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xinying Fan
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
31
|
Jin H, Sanberg PR, Henning RJ. Human umbilical cord blood mononuclear cell-conditioned media inhibits hypoxic-induced apoptosis in human coronary artery endothelial cells and cardiac myocytes by activation of the survival protein Akt. Cell Transplant 2013; 22:1637-50. [PMID: 23336598 DOI: 10.3727/096368912x661427] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We have previously demonstrated in acute myocardial infarctions that human umbilical cord blood mononuclear cells (HUCBCs), which contain hematopoietic, endothelial, and mesenchymal stem cells, reduce acute myocardial infarction size by ≥50% and preserve LV contractility. We hypothesize that the beneficial effects of HUCBCs are due to secretion of biologically active factors that activate in cardiac endothelial cells and myocytes the cell survival protein Akt. We determined by protein microarrays the growth factors and anti-inflammatory cytokines secreted by HUCBCs into culture media during 12 h of hypoxia (1% O2). We then determined by Western blots the effects of cell-free media from hypoxic-conditioned HUCBCs (HUCM) on activation of the cell survival protein Akt in human coronary artery endothelial cells and cardiac myocytes in culture during 24 h of 1% O2. We also determined in separate experiments endothelial cell and myocyte apoptosis by caspase-3 and Annexin V. In the present experiments, HUCBCs secreted multiple growth factors, anti-inflammatory cytokines, and inhibitors of metalloproteinase during normoxia and hypoxia. Human cord blood cells increased the concentration in culture media of angiopoietin, hepatocyte growth factor, interleukin-4, insulin-like growth factor, placental growth factor, vascular endothelial cell growth factor, angiogenin, and stem cell factor by 100 to >10,000% during 12 h of 1% O2 (p<0.001). HUCM, which contained these biological factors, significantly increased Akt phosphorylation/activation in coronary artery endothelial cells and cardiac myocytes subjected to 24 h of 1% O2 by more than 60% (p<0.05) and increased the antiapoptotic protein Bcl-2 expression by 34-50% in comparison with endothelial cells and myocytes treated without HUCM in 1% O2(p<0.05). HUCM also significantly decreased caspase-3 activity and decreased hypoxic endothelial cell and cardiac myocyte apoptosis by more than 40% in comparison with cells cultured without HUCM (p<0.05). Inhibition of Akt activation in endothelial cells and myocytes by the sensitive and specific antagonist API-1 during 24 h of hypoxia nearly completely prevented the beneficial effects of HUCM on inhibiting caspase-3 activity and apoptosis. We conclude that HUCBCs secrete biologically active factors during hypoxia that activate survival proteins in endothelial cells and myocytes that significantly limit apoptosis.
Collapse
Affiliation(s)
- Hua Jin
- Center for Cardiovascular Research and James A. Haley VA Medical Center, Tampa, FL, USA
| | | | | |
Collapse
|
32
|
Henning RJ, Dennis S, Sawmiller D, Hunter L, Sanberg P, Miller L. Human umbilical cord blood mononuclear cells activate the survival protein Akt in cardiac myocytes and endothelial cells that limits apoptosis and necrosis during hypoxia. Transl Res 2012; 159:497-506. [PMID: 22633101 DOI: 10.1016/j.trsl.2012.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 02/04/2012] [Accepted: 02/06/2012] [Indexed: 12/29/2022]
Abstract
We have previously reported that human umbilical cord blood mononuclear cells (HUCBC), which contain hematopoietic, mesenchymal, and endothelial stem cells, can significantly reduce acute myocardial infarction size. To determine the mechanism whereby HUCBC increase myocyte and vascular endothelial cell survival, we treated cardiac myocytes and coronary artery endothelial cells in separate experiments with HUCBC plus culture media or culture media alone and subjected the cells to 24 h of hypoxia or normoxia. We then determined in myocytes and endothelial cells activation of the cell survival protein Akt by Western blots. We also determined in these cells apoptosis by annexin V staining and necrosis by propidium iodide staining. Thereafter, we inhibited with API, a specific and sensitive Akt inhibitor, Akt activation in myocytes and endothelial cells cultured with HUCBC during hypoxia and determined cell apoptosis and necrosis. In cells cultured without HUCBC, hypoxia only slightly activated Akt. Moreover, hypoxia increased myocyte apoptosis by ≥ 226% and necrosis by 58% in comparison with myocytes in normoxia. Hypoxic treatment of endothelial cells without HUCBC increased apoptosis by 94% and necrosis by 59%. In contrast, hypoxia did not significantly affect HUCBC. Moreover, in myocyte + HUCBC cultures in hypoxia, HUCBC induced a ≥ 135% increase in myocyte phospho-Akt. Akt activation decreased myocyte apoptosis by 76% and necrosis by 35%. In endothelial cells, HUCBC increased phospho-Akt by 116%. HUCBC also decreased endothelial cell apoptosis by 58% and necrosis by 42%. Inhibition of Akt with API in myocytes and endothelial cells cultured with HUCBC during hypoxia nearly totally prevented the HUCBC-induced decrease in apoptosis and necrosis. We conclude that HUCBC can significantly decrease hypoxia-induced myocyte and endothelial cell apoptosis and necrosis by activating Akt in these cells and in this manner HUCBC can limit myocardial ischemia and injury.
Collapse
Affiliation(s)
- Robert J Henning
- James A. Haley VA Medical Center and the University of South Florida College of Medicine, Tampa, FL, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Ratajczak MZ, Zuba-Surma E, Kucia M, Poniewierska A, Suszynska M, Ratajczak J. Pluripotent and multipotent stem cells in adult tissues. Adv Med Sci 2012; 57:1-17. [PMID: 22515973 DOI: 10.2478/v10039-012-0020-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
One of the most intriguing questions in stem cell biology is whether pluripotent stem cells exist in adult tissues. Several groups of investigators employing i) various isolation protocols, ii) detection of surface markers, and iii) experimental in vitro and in vivo models, have reported the presence of cells that possess a pluripotent character in adult tissues. Such cells were assigned various operational abbreviations and names in the literature that added confusion to the field and raised the basic question of whether these are truly distinct or overlapping populations of the same primitive stem cells. Unfortunately, these cells were never characterized side-by-side to address this important issue. Nevertheless, taking into consideration their common features described in the literature, it is very likely that various investigators have described overlapping populations of developmentally early stem cells that are closely related. These different populations of stem cells will be reviewed in this paper.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Biology Program, James Graham Brown Cancer Center, University of Louisville, Kentucky, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Stem-cell Therapy for Peripheral Arterial Occlusive Disease. Eur J Vasc Endovasc Surg 2011; 42:667-75. [DOI: 10.1016/j.ejvs.2011.06.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 06/15/2011] [Indexed: 11/17/2022]
|
35
|
Venugopal JR, Prabhakaran MP, Mukherjee S, Ravichandran R, Dan K, Ramakrishna S. Biomaterial strategies for alleviation of myocardial infarction. J R Soc Interface 2011; 9:1-19. [PMID: 21900319 PMCID: PMC3223634 DOI: 10.1098/rsif.2011.0301] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
World Health Organization estimated that heart failure initiated by coronary artery disease and myocardial infarction (MI) leads to 29 per cent of deaths worldwide. Heart failure is one of the leading causes of death in industrialized countries and is expected to become a global epidemic within the twenty-first century. MI, the main cause of heart failure, leads to a loss of cardiac tissue impairment of left ventricular function. The damaged left ventricle undergoes progressive ‘remodelling’ and chamber dilation, with myocyte slippage and fibroblast proliferation. Repair of diseased myocardium with in vitro-engineered cardiac muscle patch/injectable biopolymers with cells may become a viable option for heart failure patients. These events reflect an apparent lack of effective intrinsic mechanism for myocardial repair and regeneration. Motivated by the desire to develop minimally invasive procedures, the last 10 years observed growing efforts to develop injectable biomaterials with and without cells to treat cardiac failure. Biomaterials evaluated include alginate, fibrin, collagen, chitosan, self-assembling peptides, biopolymers and a range of synthetic hydrogels. The ultimate goal in therapeutic cardiac tissue engineering is to generate biocompatible, non-immunogenic heart muscle with morphological and functional properties similar to natural myocardium to repair MI. This review summarizes the properties of biomaterial substrates having sufficient mechanical stability, which stimulates the native collagen fibril structure for differentiating pluripotent stem cells and mesenchymal stem cells into cardiomyocytes for cardiac tissue engineering.
Collapse
Affiliation(s)
- Jayarama Reddy Venugopal
- Healthcare and Energy Materials Laboratory, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, Singapore.
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Myocardial infarction is the leading cause of death among people in industrialized nations. Although the heart has some ability to regenerate after infarction, myocardial restoration is inadequate. Consequently, investigators are currently exploring the use of human embryonic stem cells (hESCs), skeletal myoblasts and adult bone marrow stem cells to limit infarct size. hESCs are pluripotent cells that can regenerate myocardium in infarcted hearts, attenuate heart remodeling and contribute to left ventricle (LV) systolic force development. Since hESCs can form heart teratomas, investigators are differentiating hESCs toward cardiac progenitor cells prior to transplantation into hearts. Large quantities of hESCs cardiac progenitor cells, however, must be generated, immune rejection must be prevented and grafts must survive over the long term to significantly improve myocardial performance. Transplanted autologous skeletal myoblasts can survive in infarcted myocardium in small numbers, proliferate, differentiate into skeletal myofibers and increase the LV ejection fraction. These cells, however, do not form electromechanical connections with host cardiomyocytes. Consequently, electrical re-entry can occur and cause cardiac arrhythmias. Autologous bone marrow mononuclear cells contain hematopoietic and mesenchymal stem cells. In several meta-analyses, patients with coronary disease who received autologous bone marrow cells by intracoronary injection show significant 3.7% (range: 1.9-5.4%) increases in LV ejection fraction, decreases in LV end-systolic volume of -4.8 ml (range: -1.4 to -8.2 ml) and reductions in infarct size of 5.5% (-1.9 to -9.1%), without experiencing arrhythmias. Bone marrow cells appear to release biologically active factors that limit myocardial damage. Unfortunately, bone marrow cells from patients with chronic diseases propagate poorly and can die prematurely. Substantial challenges must be addressed and resolved to advance the use of stem cells in cardiac repair including identifying the optimal stem cell(s) that permit transplantation without requirements for host immune suppression; timing of stem cell transplantation that maximizes chemoattraction of stem cells to infarcts; and determining the optimal technique for injecting stem cells for cardiac repair. Techniques must be developed to enhance survival and propagation of stem cells in the myocardium. These studies will require close cooperation and interaction of scientists and clinicians. Cell-based cardiac repair in the 21st century will offer new hope for millions of patients worldwide with myocardial infarctions who, otherwise, would suffer from the relentless progression of heart disease to heart failure and death.
Collapse
Affiliation(s)
- Robert J Henning
- James A. Haley VA Hospital/University of South Florida College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
37
|
Klopsch C, Donndorf P, Kaminski A, Ma N, Steinhoff G. Zellquellen für kardiovaskuläres Tissue Engineering. Chirurg 2011; 82:295-302. [DOI: 10.1007/s00104-010-2030-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Arien-Zakay H, Lazarovici P, Nagler A. Tissue regeneration potential in human umbilical cord blood. Best Pract Res Clin Haematol 2011; 23:291-303. [PMID: 20837341 DOI: 10.1016/j.beha.2010.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative medicine is the process of creating functional tissue with the aid of stem cells, to repair loss of organ function. Possible targets for regenerative medicine include orthopaedic, cardiac, hepatic, pancreatic and central nervous system (CNS) applications. Umbilical cord blood (CB) has established itself as a legitimate source for haematopoietic stem cell transplantation. It is also considered an accessible and less immunogenic source for mesenchymal, unrestricted somatic and for other stem cells with pluri/multipotent properties. The latter are capable of differentiating into a wide variety of cell types including bone, cartilage, cardiomyocytes and neural. They also possess protective abilities that may contribute to tissue repair even if in vitro differentiation is excluded. In view of the absence of treatment for many devastating diseases, the elucidation of non-haematopoietic applications for CB will facilitate the development of pioneering relevant cell therapy approaches. This review focusses on current studies using human CB-derived cells for regenerative medicine.
Collapse
|
39
|
Cord blood transplantation and stem cell regenerative potential. Exp Hematol 2011; 39:393-412. [PMID: 21238533 DOI: 10.1016/j.exphem.2011.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 01/06/2011] [Accepted: 01/08/2011] [Indexed: 02/06/2023]
Abstract
The past 20 years of experience with umbilical cord blood transplantation have demonstrated that cord blood is effective in the treatment of a spectrum of diseases, including hematological malignancies, bone marrow failure, hemoglobinopathies, and inborn errors of metabolism. Cord blood can be obtained with ease and then safely cryopreserved for either public or private use without loss of viability. As compared to other unrelated donor cell sources, cord blood transplantation allows for greater human leukocyte antigen disparity without a corresponding increase in graft-vs.-host disease. Moreover, cord blood has a lower risk of transmitting infections by latent viruses and is less likely to carry somatic mutations than other adult cells. Recently, multiple populations of stem cells with primitive stem cell properties have been identified from cord blood. Meanwhile, there is an increasing interest in applying cord blood mononuclear cells or enriched stem cell populations to regenerative therapies. Accumulating evidence has suggested functional improvements after cord blood transplantation in various animal models for treatments of cardiac infarction, diabetes, neurological diseases, etc. In this review, we will summarize the most recent updates on clinical applications of cord blood transplantation and the promises and limitations of cell-based therapies for tissue repair and regeneration.
Collapse
|
40
|
Lupu M, Khalil M, Andrei E, Iordache F, Pfannkuche K, Neef K, Georgescu A, Buzila C, Brockmeier K, Maniu H, Hescheler J. Integration Properties of Wharton’s Jelly-derived Novel Mesenchymal Stem Cells into Ventricular Slices of Murine Hearts. Cell Physiol Biochem 2011; 28:63-76. [DOI: 10.1159/000331714] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2011] [Indexed: 02/06/2023] Open
|
41
|
Senegaglia AC, Barboza LA, Dallagiovanna B, Aita CAM, Hansen P, Rebelatto CLK, Aguiar AM, Miyague NI, Shigunov P, Barchiki F, Correa A, Olandoski M, Krieger MA, Brofman PRS. Are purified or expanded cord blood-derived CD133+ cells better at improving cardiac function? Exp Biol Med (Maywood) 2010; 235:119-29. [PMID: 20404026 DOI: 10.1258/ebm.2009.009194] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Endothelial progenitor cells (EPCs), which express the CD133 marker, can differentiate into mature endothelial cells (ECs) and create new blood vessels. Normal angiogenesis is unable to repair the injured tissues that result from myocardial infarction (MI). Patients who have high cardiovascular risks have fewer EPCs and their EPCs exhibit greater in vitro senescence. Human umbilical cord blood (HUCB)-derived EPCs could be an alternative to rescue impaired stem cell function in the sick and elderly. The aim of this study was to purify HUCB-derived CD133(+) cells, expand them in vitro and evaluate the efficacy of the purified and expanded cells in treating MI in rats. CD133(+) cells were selected for using CD133-coupled magnetic microbeads. Purified cells stained positive for EPC markers. The cells were expanded and differentiated in media supplemented with fetal calf serum and basic fibroblast growth factor, insulin-like growth factor-I and vascular endothelial growth factor (VEGF). Differentiation was confirmed by lack of staining for EPC markers. These expanded cells exhibited increased expression of mature EC markers and formed tubule-like structures in vitro. Only the expanded cells expressed VEGF mRNA. Cells were expanded up to 70-fold during 60 days of culture, and they retained their functional activity. Finally, we evaluated the therapeutic potential of purified and expanded CD133(+) cells in treating MI by intramyocardially injecting them into a rat model of MI. Rats were divided into three groups: A (purified CD133(+) cells-injected); B (expanded CD133(+) cells-injected) and C (saline buffer-injected). We observed a significant improvement in left ventricular ejection fraction for groups A and B. In summary, CD133(+) cells can be purified from HUCB, expanded in vitro without loosing their biological activity, and both purified and expanded cells show promising results for use in cellular cardiomyoplasty. However, further pre-clinical testing should be performed to determine whether expanded CD133(+) cells have any clinical advantages over purified CD133(+) cells.
Collapse
Affiliation(s)
- Alexandra C Senegaglia
- Pontifícia Universidade Católica do Paraná, Institute for Health and Biological Sciences, Rua Imaculada Conceição, 1155 Curitiba, Paraná, 80215901, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Henning RJ, Aufman J, Shariff M, Sawmiller D, DeLostia V, Sanberg P, Morgan M. Human umbilical cord blood mononuclear cells decrease fibrosis and increase cardiac function in cardiomyopathy. Regen Med 2010; 5:45-54. [PMID: 20017693 DOI: 10.2217/rme.09.71] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
AIMS We investigated whether human umbilical cord blood mononuclear cells (HUCBC) can limit progressive cardiomyopathy in TO2 hamsters. MATERIALS & METHODS A total of 22 TO2 1-month-old hamsters were treated with intramyocardial HUCBC, 4 x 10(6) in Isolyte, and 23 TO2 1-month-old hamsters were treated with intramyocardial Isolyte. A total of 16 1-month-old F1B hamsters served as controls and received intramyocardial Isolyte. Echocardiograms were performed on all hamsters prior to and monthly after treatment for 6 months. Heart tissues were then stained with hematoxylin and eosin, Masson's Trichrome and human leukocyte antibody. RESULTS In F1B hamsters, left ventricular fractional shortening (FS) and ejection fractions (EF) did not significantly decrease over 6 months. By contrast, in Isolyte-treated TO2 hamsters, FS decreased from 56.2 +/- 1.0% to 19.7 +/- 3.2% and EF decreased from 89.5 +/- 1.4% to 44.9 +/- 5.9% at 6 months (both p < 0.0001). The FS and EF in HUCBC-treated TO2 hamsters also progressively decreased over 6 months but the changes were more gradual, especially during the first month after HUCBC treatment when FS was 52.0 +/- 1.5% and EF was 89.5 +/- 1.4%, which was not significantly different from the FS and EF in the F1B hamsters. Moreover, in the HUCBC-treated hamsters, the FS and EF were 20-30% greater than FS and EF in Isolyte TO2 hamsters at 3 and 5 months (p < 0.01). In Isolyte-treated TO2 hamsters at 6-7 months, fibrosis involved 30.0 +/- 5.0% of left ventricle and 35.0 +/- 5.0% of septum. By contrast, in HUCBC-treated hamsters, fibrosis involved only 6.5 +/- 2.3% of the left ventricle and 6.3 +/- 1.8% of septum (p < 0.05). The average number of blood vessels per myocardial microscopic field in HUCBC-treated hearts was 53.5 +/- 0.8 versus 46.2 +/- 3.0 in Isolyte-treated TO2 hearts (p < 0.05). CONCLUSION HUCBC, when given as a single intramyocardial injection, can limit fibrosis and increase heart function over the short term in TO2 hamsters with cardiomyopathy.
Collapse
Affiliation(s)
- Robert J Henning
- Center for Cardiovascular Research, James A Haley Hospital/University of South Florida, 13000 Bruce B. Downs Blvd, 111, Tampa, Florida 33612, FL, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Gopinath S, Vanamala SK, Gondi CS, Rao JS. Human umbilical cord blood derived stem cells repair doxorubicin-induced pathological cardiac hypertrophy in mice. Biochem Biophys Res Commun 2010; 395:367-72. [PMID: 20382121 DOI: 10.1016/j.bbrc.2010.04.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 04/02/2010] [Indexed: 12/31/2022]
Abstract
In the present study, we investigated the cardiomyogenic potential of human umbilical cord blood (hUCB)-derived stem cells and whether stem cell treatment repairs the pathological hypertrophy induced by doxorubicin (DOX) in cultured neonatal rat cardiomyocytes (NRCM) and in mouse hearts. hUCB, which were labeled with cell tracker dye, were co-cultured with isolated NRCM in vitro. After 48h of incubation, the red stained hUCB cells (30%) contracted rhythmically and synchronously (physical examination). These differentiated hUCB also expressed cardiac specific alpha-actinin and showed diffused expression of connexin 43 and N-cadherin, thereby suggesting a tight electrical coupling among hUCB cells and myocytes. When co-cultured, hUCB also reversed the pathological effects induced by DOX in NRCM and in mice as seen by RT-PCR, immunoblot analysis and immunocytochemistry. hUCB migrated and integrated into the hearts of mice that were treated with DOX after intravenous injection and reversed the expression of pathological hypertrophic markers induced by DOX in mice. Further, we observed a shift from pathological hypertrophy towards physiological hypertrophy by hUCB in DOX-challenged mice. hUCB treatment in mice decreased DOX-induced increase of heart weight to body mass ratio and fibrosis. Taken together, these findings suggest the potential therapeutic use of hUCB in reversing heart failure conditions.
Collapse
Affiliation(s)
- Sreelatha Gopinath
- Department of Cancer Biology & Pharmacology, University of Illinois - College of Medicine at Peoria, Peoria, IL 61656, USA
| | | | | | | |
Collapse
|
44
|
Pinho-Ribeiro V, Maia ACV, Werneck-de-Castro JPS, Oliveira PF, Goldenberg RCS, Carvalho ACCD. Human umbilical cord blood cells in infarcted rats. Braz J Med Biol Res 2010; 43:290-296. [PMID: 20401437 DOI: 10.1590/s0100-879x2010007500007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 01/26/2010] [Indexed: 02/08/2023] Open
Abstract
Therapy with bone marrow-derived cells has been used in ischemic patients with reported success. The aim of this study was to determine the therapeutic efficacy of fresh and frozen human umbilical cord blood cells (hUCB) in Wistar rats submitted to permanent occlusion of the left coronary artery. Three hours after myocardial infarction, 2 x 10(7) hUCB cells or vehicle were administered by intramyocardial injection. The animals were divided into five groups: control (N = 10), sham operated (N = 10), infarcted that received vehicle (N = 9), infarcted treated with cryopreserved hUCB (N = 7), and infarcted treated with fresh hUCB (N = 5). Cardiac function was evaluated by electrocardiogram (ECG) and echocardiogram (ECHO) before cell therapy, and by ECG, ECHO, cardiopulmonary test, and left ventricular pressure measurements 3 weeks later. After 3 weeks, both groups treated with hUCB still had Q wave present in L1, âQRS >90 degrees and reduced shortening fraction (less than 50%). In addition, cardiac indexes of left ventricular contractility and relaxation were 5484 +/- 875 and -4032 +/- 643 mmHg (cryopreserved hUCB) and 4585 +/- 955 and -2862 +/- 590 mmHg (fresh hUCB), respectively. These values were not statistically different from those of saline-treated animals. Cardiopulmonary exercise test profile was typical of infarcted hearts; exercise time was about 14 min and maximal VO2 was 24.77 +/- 5.00 mL.kg-1.min-1. These data show that hUCB therapy did not improve the cardiac function of infarcted animals or prevent cardiac remodeling.
Collapse
Affiliation(s)
- V Pinho-Ribeiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brasil
| | | | | | | | | | | |
Collapse
|
45
|
Zech NH, Broer N, Ribitsch I, Zech MH, Broer KH, Ertan K, Preisegger KH. The rationale behind collecting umbilical cord blood. J Turk Ger Gynecol Assoc 2010; 11:99-101. [PMID: 24591908 DOI: 10.5152/jtgga.2010.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Accepted: 12/31/2009] [Indexed: 11/22/2022] Open
Abstract
Umbilical cord blood (UCB) is an increasingly important and rich source of stem cells. These cells can be used for the treatment of many diseases, including cancers and immune and genetic disorders. For patients for whom no suitable related donor is available, this source of hematopoietic stem cells offers substantial advantages, notably the relative ease of procurement, the absence of risk to the donor, the small likelihood of transmitting clinically important infections, the low risk of severe graft-versus-host disease (GVHD) and the rapid availability of placental blood for transplantation centers. Even though almost 80 diseases are treatable with cord blood stem cells, 97 percent of cord blood is still disposed of after birth and lost for patients in need! To improve availability of stem cells to a broader community, efforts should be undertaken to collect cord blood and expectant parents should be properly informed of their options with regard to cord blood banking.
Collapse
Affiliation(s)
- Nicolas H Zech
- Ivf Centers Prof. Zech, Bregenz, Austria and Department For Obstetrics and Gynecology, Unit of Gynecological Endocrinology and Reproductive Medicine, University of Graz, Austria
| | - Nikolas Broer
- Yale University School of Medicine, Usa, and Center For Reproductive Medicine, Cologne, Germany
| | - Iris Ribitsch
- Vivocell Biosolutions Gmbh, Graz, Austria and Langenfeld/Düsseldorf, Germany
| | - Mathias H Zech
- Ivf Centers Prof. Zech, Bregenz, Austria and Department For Obstetrics and Gynecology, Unit of Gynecological Endocrinology and Reproductive Medicine, University of Graz, Austria
| | - Karl-Heinz Broer
- Vivocell Biosolutions Gmbh, Graz, Austria and Langenfeld/Düsseldorf, Germany
| | - Kubilay Ertan
- Klinikum Leverkusen, Department of Obstretics and Gynecology, Leverkusen, Germany
| | | |
Collapse
|
46
|
Hossne NA, Invitti AL, Buffolo E, Azevedo S, Rodrigues de Oliveira JS, Stolf NG, Cruz LE, Sanberg PR. Refractory angina cell therapy (ReACT) involving autologous bone marrow cells in patients without left ventricular dysfunction: a possible role for monocytes. Cell Transplant 2009; 18:1299-310. [PMID: 20149298 DOI: 10.3727/096368909x484671] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Autologous bone marrow mononuclear cell (BMMC) transplantation has emerged as a potential therapeutic option for refractory angina patients. Previous studies have shown conflicting myocardium reperfusion results. The present study evaluated safety and efficacy of CellPraxis Refractory Angina Cell Therapy Protocol (ReACT), in which a specific BMMC formulation was administered as the sole therapy for these patients. The phase I/IIa noncontrolled, open label, clinical trial, involved eight patients with refractory angina and viable ischemic myocardium, without left ventricular dysfunction and who were not suitable for conventional myocardial revascularization. ReACT is a surgical procedure involving a single series of multiple injections (40-90 injections, 0.2 ml each) into ischemic areas of the left ventricle. Primary endpoints were Canadian Cardiovascular Society Angina Classification (CCSAC) improvement at 18 months follow-up and myocardium ischemic area reduction (assessed by scintigraphic analysis) at 12 months follow-up, in correlation with a specific BMMC formulation. Almost all patients presented progressive improvement in angina classification beginning 3 months (p = 0.008) postprocedure, which was sustained at 18 months follow-up (p = 0.004), as well as objective myocardium ischemic area reduction at 12 months (decrease of 84.4%, p < 0.004). A positive correlation was found between monocyte concentration and CCSAC improvement (r = -0.759, p < 0.05). Improvement in CCSAC, followed by correlated reduction in scintigraphic myocardium ischemic area, strongly suggests neoangiogenesis as the main stem cell action mechanism. The significant correlation between number of monocytes and improvement strongly supports a cell-related effect of ReACT. ReACT appeared safe and effective.
Collapse
Affiliation(s)
- Nelson Americo Hossne
- Cardiovascular Surgery Division, Surgery Department, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Stem cells contribute to innate healing and harbor a promising role for regenerative medicine. Stem cell banking through long-term storage of different stem cell platforms represents a fundamental source to preserve original features of stem cells for patient-specific clinical applications. Stem cell research and clinical translation constitute fundamental and indivisible modules catalyzed through biobanking activity, generating a return of investment.
Collapse
|
48
|
Yerebakan C, Sandica E, Prietz S, Klopsch C, Ugurlucan M, Kaminski A, Abdija S, Lorenzen B, Boltze J, Nitzsche B, Egger D, Barten M, Furlani D, Ma N, Vollmar B, Liebold A, Steinhoff G. Autologous umbilical cord blood mononuclear cell transplantation preserves right ventricular function in a novel model of chronic right ventricular volume overload. Cell Transplant 2009; 18:855-68. [PMID: 19500473 DOI: 10.3727/096368909x471170] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We aimed to evaluate the feasibility and efficacy of autologous umbilical cord blood mononuclear cell (UCMNC) transplantation on right ventricular (RV) function in a novel model of chronic RV volume overload. Four-month-old sheep (n = 20) were randomized into cell (n = 10) and control groups (n = 10). After assessment of baseline RV function by the conductance catheter method, a transannular patch (TAP) was sutured to the right ventricular outflow tract (RVOT). Following infundibulotomy the ring of the pulmonary valve was transected without cardiopulmonary bypass. UCMNC implantation (8.22 +/- 6.28 x 10(7)) in the cell group and medium injection in the control group were performed into the RV myocardium around the TAP. UCMNCs were cultured for 2 weeks after fluorescence-activated cell sorting (FACS) analysis for CD34 antigen. Transthoracic echocardiography (TTE) and computed tomography were performed after 6 weeks and 3 months, respectively. RV function was assessed 3 months postoperatively before the hearts were excised for immunohistological examinations. FACS analysis revealed 1.2 +/- 0.22% CD34(+) cells within the isolated UCMNCs from which AcLDL(+) endothelial cells were cultured in vitro. All animals survived surgery. TTE revealed grade II-III pulmonary regurgitation in both groups. Pressure-volume loops under dobutamine stress showed significantly improved RV diastolic function in the cell group (dP/dt(min): p = 0.043; E(ed): p = 0.009). CD31 staining indicated a significantly enhanced number of microvessels in the region of UCMNC implantation in the cell group (p < 0.001). No adverse tissue changes were observed. TAP augmentation and pulmonary annulus distortion without cardiopulmonary bypass constitutes a valid large animal model mimicking the surgical repair of tetralogy of Fallot. Our results indicate that the chronically volume-overloaded RV profits from autologous UCMNC implantation by enhanced diastolic properties with a probable underlying mechanism of increased angiogenesis.
Collapse
Affiliation(s)
- Can Yerebakan
- Department of Cardiac Surgery, Medical Faculty, University of Rostock, Rostock, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Ratajczak MZ, Kucia M, Ratajczak J, Zuba-Surma EK. A multi-instrumental approach to identify and purify very small embryonic like stem cells (VSELs) from adult tissues. Micron 2009; 40:386-93. [DOI: 10.1016/j.micron.2008.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 09/27/2008] [Indexed: 11/17/2022]
|
50
|
Hall AA, Guyer AG, Leonardo CC, Ajmo CT, Collier LA, Willing AE, Pennypacker KR. Human umbilical cord blood cells directly suppress ischemic oligodendrocyte cell death. J Neurosci Res 2009; 87:333-41. [PMID: 18924174 DOI: 10.1002/jnr.21857] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Previous reports have shown that human umbilical cord blood cells (HUCBCs) administered intravenously 48 hr following middle cerebral artery occlusion reduce infarct area and behavioral deficits of rodents. This cellular therapy is potently neuroprotective and antiinflammatory. This study investigates the effect of HUCBC treatment on white matter injury and oligodendrocyte survival in a rat model of ischemia. Intravenous infusion of 10(6) HUCBCs 48 hr poststroke reduced the amount of white matter damage in vivo as seen by quantification of myelin basic protein staining in tissue sections. To determine whether HUCBC treatment was protective via direct effects on oligodendrocytes, cultured oligodendrocytes were studied in an in vitro model of oxygen glucose deprivation. Active caspase 3 immunohistochemistry and the lactate dehydrogenase assay for cytotoxicity were used to determine that HUCBCs provide protection to oligodendrocytes in vitro. Based on these results, it is likely that HUCBC administration directly protects oligodendrocytes and white matter. This effect is likely to contribute to the increased behavioral recovery observed with HUCBC therapy.
Collapse
Affiliation(s)
- A A Hall
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | | | | | | | | | | |
Collapse
|