201
|
Projahn D, Simsekyilmaz S, Singh S, Kanzler I, Kramp BK, Langer M, Burlacu A, Bernhagen J, Klee D, Zernecke A, Hackeng TM, Groll J, Weber C, Liehn EA, Koenen RR. Controlled intramyocardial release of engineered chemokines by biodegradable hydrogels as a treatment approach of myocardial infarction. J Cell Mol Med 2014; 18:790-800. [PMID: 24512349 PMCID: PMC4119385 DOI: 10.1111/jcmm.12225] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/03/2013] [Indexed: 12/26/2022] Open
Abstract
Myocardial infarction (MI) induces a complex inflammatory immune response, followed by the remodelling of the heart muscle and scar formation. The rapid regeneration of the blood vessel network system by the attraction of hematopoietic stem cells is beneficial for heart function. Despite the important role of chemokines in these processes, their use in clinical practice has so far been limited by their limited availability over a long time-span in vivo. Here, a method is presented to increase physiological availability of chemokines at the site of injury over a defined time-span and simultaneously control their release using biodegradable hydrogels. Two different biodegradable hydrogels were implemented, a fast degradable hydrogel (FDH) for delivering Met-CCL5 over 24 hrs and a slow degradable hydrogel (SDH) for a gradual release of protease-resistant CXCL12 (S4V) over 4 weeks. We demonstrate that the time-controlled release using Met-CCL5-FDH and CXCL12 (S4V)-SDH suppressed initial neutrophil infiltration, promoted neovascularization and reduced apoptosis in the infarcted myocardium. Thus, we were able to significantly preserve the cardiac function after MI. This study demonstrates that time-controlled, biopolymer-mediated delivery of chemokines represents a novel and feasible strategy to support the endogenous reparatory mechanisms after MI and may compliment cell-based therapies.
Collapse
Affiliation(s)
- Delia Projahn
- Institute for Molecular Cardiovascular Research (IMCAR), Medical Faculty, RWTH Aachen UniversityAachen, Germany
- Institute for Cardiovascular Prevention (IPEK), University Hospital of the LMU MunichMunich, Germany
| | - Sakine Simsekyilmaz
- Institute for Molecular Cardiovascular Research (IMCAR), Medical Faculty, RWTH Aachen UniversityAachen, Germany
| | - Smriti Singh
- Interactive Material Research - DWI an der RWTH Aachen e.V and Institute for Technical and Macromolecular ChemistryAachen, Germany
| | - Isabella Kanzler
- Institute for Molecular Cardiovascular Research (IMCAR), Medical Faculty, RWTH Aachen UniversityAachen, Germany
| | - Birgit K Kramp
- Institute for Cardiovascular Prevention (IPEK), University Hospital of the LMU MunichMunich, Germany
| | - Marcella Langer
- Institute for Cardiovascular Prevention (IPEK), University Hospital of the LMU MunichMunich, Germany
| | - Alexandrina Burlacu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian AcademyBucharest, Romania
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell BiologyAachen, Germany
| | - Doris Klee
- Interactive Material Research - DWI an der RWTH Aachen e.V and Institute for Technical and Macromolecular ChemistryAachen, Germany
| | - Alma Zernecke
- Department of Vascular Surgery, Klinikum rechts der Isar, Technical University MunichMunich, Germany
| | - Tilman M Hackeng
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UniversityMaastricht, The Netherlands
| | - Jürgen Groll
- Department and Chair of Functional Materials in Medicine and Dentistry, University of WürzburgWürzburg, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), University Hospital of the LMU MunichMunich, Germany
- Munich Heart AllianceMunich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UniversityMaastricht, The Netherlands
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research (IMCAR), Medical Faculty, RWTH Aachen UniversityAachen, Germany
| | - Rory R Koenen
- Institute for Cardiovascular Prevention (IPEK), University Hospital of the LMU MunichMunich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht UniversityMaastricht, The Netherlands
- *Correspondence to: Rory R. KOENEN, Ph.D., Institute for Cardiovascular Prevention (IPEK), University Hospital of the Ludwig-Maximilians-University Munich, Pettenkoferstraße 9, Munich 80336, Germany. Tel.: +49 89 51604672 Fax: +49 89 51604352 E-mail:
| |
Collapse
|
202
|
Dash SN, Dash NR, Guru B, Mohapatra PC. Towards Reaching the Target: Clinical Application of Mesenchymal Stem Cells for Diabetic Foot Ulcers. Rejuvenation Res 2014; 17:40-53. [DOI: 10.1089/rej.2013.1467] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
| | - Nihar Ranjan Dash
- Department of Biochemistry, Apollo Hospitals Bhubaneswar, Odisha. India
| | | | | |
Collapse
|
203
|
Abstract
Extensive studies in experimental animal heart models and patients have shown the promise of bone marrow cell (BMC) transplantation as an alternative strategy to the conventional treatment modalities for cardiac repair. 'Stemness' of BMC to adopt cardiac phenotype, their potential as carriers of exogenous therapeutic genes and an inherent ability to express growth factors and cytokines to exert paracrine effects have been especially focused until recently. These findings suggest that locally delivered BMCs are capable of regenerating de novo myocardium. Others have shown that extensive neovascularization due to paracrine effects of the engrafted cells resulted in improved regional blood flow and reduced infarct size. Despite initial success, there are multiple fundamental issues that remain contentious. Indeed, resolving these issues will optimize future heart cell therapy protocols to achieve better prognosis in the clinical settings. This review is a concise, in-depth and critical appreciation of the role of BMCs in heart cell therapy and builds a conceptual framework to elaborate their significance as a possible source of donor cells. Moreover, it discusses the current status of BMC transplantation as a clinical modality and the relevant issues confronting this approach in light of the published data with clinical relevance.
Collapse
Affiliation(s)
- Husnain Kh Haider
- Department of Pathology and Laboratory of Medicine, 231-Albert Sabinway, Cinncinati, OH 45267-0529, USA.
| |
Collapse
|
204
|
Abstract
Cardiovascular disease remains the single greatest cause of death in the Western world, claiming more lives in the USA than the next four leading causes combined. Among these diseases, the incidence of heart failure continues to rise at a staggering rate. Recent advances in medical and device therapies have dramatically improved both the survival and quality of life of many of these patients; however, limited strategies are available to address the central pathophysiology underlying the development of heart failure, namely, the loss of functional cardiomyocytes. Therefore, one recent strategy has been the development of cell-based therapies, aiming towards the replacement of injured or lost cardiomyocytes and thereby improved cardiac function. In this review, we will examine the cell types undergoing investigation as potential cell-based therapies and provide an overview of current clinical trials utilizing cell-based therapeutic approaches in patients with heart disease.
Collapse
Affiliation(s)
- Regina L Sohn
- Cardiac Muscle Research Laboratory, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
205
|
Abstract
Therapeutic angiogenesis offers promise as a novel treatment for ischemic heart disease, particularly for patients who are not candidates for current methods of revascularization. The goal of treatment is both relief of symptoms of coronary artery disease and improvement of cardiac function by increasing perfusion to the ischemic region. Protein-based therapy with cytokines including vascular endothelial growth factor and fibroblast growth factor demonstrated functionally significant angiogenesis in several animal models. However, clinical trials have yielded largely disappointing results. The attenuated angiogenic response seen in clinical trials of patients with coronary artery disease may be due to multiple factors including endothelial dysfunction, particularly in the context of advanced atherosclerotic disease and associated comorbid conditions, regimens of single agents, as well as inefficiencies of current delivery methods. Gene therapy has several advantages over protein therapy and recent advances in gene transfer techniques have improved the feasibility of this approach. The safety and tolerability of therapeutic angiogenesis by gene transfer has been demonstrated in phase I clinical trials. The utility of therapeutic angiogenesis by gene transfer as a treatment option for ischemic cardiovascular disease will be determined by adequately powered, randomized, placebo-controlled Phase II and III clinical trials. Cell-based therapies offer yet another approach to therapeutic angiogenesis. Although it is a promising therapeutic strategy, additional preclinical studies are warranted to determine the optimal cell type to be administered, as well as the optimal delivery method. It is likely the optimal treatment will involve multiple agents as angiogenesis is a complex process involving a large cascade of cytokines, as well as cells and extracellular matrix, and administration of a single factor may be insufficient. The promise of therapeutic angiogenesis as a novel treatment for no-option patients should be approached with cautious optimism as the field progresses.
Collapse
Affiliation(s)
- Audrey Rosinberg
- Division of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center, 110 Francis Street, Suite 2A, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
206
|
Yan J, Tie G, Xu TY, Cecchini K, Messina LM. Mesenchymal stem cells as a treatment for peripheral arterial disease: current status and potential impact of type II diabetes on their therapeutic efficacy. Stem Cell Rev Rep 2014; 9:360-72. [PMID: 23475434 DOI: 10.1007/s12015-013-9433-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs), due to their paracrine, transdifferentiation, and immunosuppressive effects, hold great promise as a therapy for peripheral arterial disease. Diabetes is an important risk factor for peripheral arterial disease; however, little is known of how type II diabetes affects the therapeutic function of MSCs. This review summarizes the current status of preclinical and clinical studies that have been performed to determine the efficacy of MSCs in the treatment of peripheral arterial disease. We also present findings from our laboratory regarding the impact of type II diabetes on the therapeutic efficacy of MSCs neovascularization after the induction of hindlimb ischemia. In our studies, we documented that experimental type II diabetes in db/db mice impaired MSCs' therapeutic function by favoring their differentiation towards adipocytes, while limiting their differentiation towards endothelial cells. Moreover, type II diabetes impaired the capacity of MSCs to promote neovascularization in the ischemic hindlimb. We further showed that these impairments of MSC function and multipotency were secondary to hyperinsulinemia-induced, Nox4-dependent oxidant stress in db/db MSCs. Should human MSCs display similar oxidant stress-induced impairment of function, these findings might permit greater leverage of the potential of MSC transplantation, particularly in the setting of diabetes or other cardiovascular risk factors, as well as provide a therapeutic approach by reversing the oxidant stress of MSCs prior to transplantation.
Collapse
Affiliation(s)
- Jinglian Yan
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | | | | | | | |
Collapse
|
207
|
The role of bioactive lipids in stem cell mobilization and homing: novel therapeutics for myocardial ischemia. BIOMED RESEARCH INTERNATIONAL 2014; 2014:653543. [PMID: 24672794 PMCID: PMC3930186 DOI: 10.1155/2014/653543] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/13/2013] [Accepted: 10/11/2013] [Indexed: 11/25/2022]
Abstract
Despite significant advances in medical therapy and interventional strategies, the prognosis of millions of patients with acute myocardial infarction (AMI) and ischemic heart disease (IHD) remains poor. Currently, short of heart transplantation with all of its inherit limitations, there are no available treatment strategies that replace the infarcted myocardium. It is now well established that cardiomyocytes undergo continuous renewal, with contribution from bone marrow (BM)-derived stem/progenitor cells (SPCs). This phenomenon is upregulated during AMI by initiating multiple innate reparatory mechanisms through which BMSPCs are mobilized towards the ischemic myocardium and contribute to myocardial regeneration. While a role for the SDF-1/CXCR4 axis in retention of BMSPCs in bone marrow is undisputed, its exclusive role in their mobilization and homing to a highly proteolytic microenvironment, such as the ischemic/infarcted myocardium, is currently being challenged. Recent evidence suggests a pivotal role for bioactive lipids in the mobilization of BMSPCs at the early stages following AMI and their homing towards ischemic myocardium. This review highlights the recent advances in our understanding of the mechanisms of stem cell mobilization, provides newer evidence implicating bioactive lipids in BMSPC mobilization and differentiation, and discusses their potential as therapeutic agents in the treatment of IHD.
Collapse
|
208
|
Abotalib Z. WITHDRAWN: Importance of cord blood stem cells in regenerative medicine. Saudi J Biol Sci 2014. [DOI: 10.1016/j.sjbs.2013.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
209
|
Poole J, Mavromatis K, Binongo JN, Khan A, Li Q, Khayata M, Rocco E, Topel M, Zhang X, Brown C, Corriere MA, Murrow J, Sher S, Clement S, Ashraf K, Rashed A, Kabbany T, Neuman R, Morris A, Ali A, Hayek S, Oshinski J, Yoon YS, Waller EK, Quyyumi AA. Effect of progenitor cell mobilization with granulocyte-macrophage colony-stimulating factor in patients with peripheral artery disease: a randomized clinical trial. JAMA 2013; 310:2631-9. [PMID: 24247554 PMCID: PMC9136711 DOI: 10.1001/jama.2013.282540] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Many patients with peripheral artery disease (PAD) have walking impairment despite therapy. Experimental studies in animals demonstrate improved perfusion in ischemic hind limb after mobilization of bone marrow progenitor cells (PCs), but whether this is effective in patients with PAD is unknown. OBJECTIVE To investigate whether therapy with granulocyte-macrophage colony-stimulating factor (GM-CSF) improves exercise capacity in patients with intermittent claudication. DESIGN, SETTING, AND PARTICIPANTS In a phase 2 double-blind, placebo-controlled study, 159 patients (median [SD] age, 64 [8] years; 87% male, 37% with diabetes) with intermittent claudication were enrolled at medical centers affiliated with Emory University in Atlanta, Georgia, between January 2010 and July 2012. INTERVENTIONS Participants were randomized (1:1) to received 4 weeks of subcutaneous injections of GM-CSF (leukine), 500 μg/day 3 times a week, or placebo. Both groups were encouraged to walk to claudication daily. MAIN OUTCOMES AND MEASURES The primary outcome was peak treadmill walking time (PWT) at 3 months. Secondary outcomes were PWT at 6 months and changes in circulating PC levels, ankle brachial index (ABI), and walking impairment questionnaire (WIQ) and 36-item Short-Form Health Survey (SF-36) scores. RESULTS Of the 159 patients randomized, 80 were assigned to the GM-CSF group. The mean (SD) PWT at 3 months increased in the GM-CSF group from 296 (151) seconds to 405 (248) seconds (mean change, 109 seconds [95% CI, 67 to 151]) and in the placebo group from 308 (161) seconds to 376 (182) seconds (change of 56 seconds [95% CI, 14 to 98]), but this difference was not significant (mean difference in change in PWT, 53 seconds [95% CI, -6 to 112], P = .08). At 3 months, compared with placebo, GM-CSF improved the physical functioning subscore of the SF-36 questionnaire by 11.4 (95% CI, 6.7 to 16.1) vs 4.8 (95% CI, -0.1 to 9.6), with a mean difference in change for GM-CSF vs placebo of 7.5 (95% CI, 1.0 to 14.0; P = .03). Similarly, the distance score of the WIQ improved by 12.5 (95% CI, 6.4 to 18.7) vs 4.8 (95% CI, -0.2 to 9.8) with GM-CSF compared with placebo (mean difference in change, 7.9 [95% CI, 0.2 to 15.7], P = .047). There were no significant differences in the ABI, WIQ distance and speed scores, claudication onset time, or mental or physical component scores of the SF-36 between the groups. CONCLUSIONS AND RELEVANCE Therapy with GM-CSF 3 times a week did not improve treadmill walking performance at the 3-month follow-up. The improvements in some secondary outcomes with GM-CSF suggest that it may warrant further study in patients with claudication. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT01041417.
Collapse
Affiliation(s)
- Joseph Poole
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Kreton Mavromatis
- Division of Cardiology, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - José N Binongo
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ali Khan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Qunna Li
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Mohamed Khayata
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Elizabeth Rocco
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Matthew Topel
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Xin Zhang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Charlene Brown
- Division of Cardiology, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Matthew A Corriere
- Division of Cardiology, Atlanta Veterans Affairs Medical Center, Decatur, Georgia6Division of Vascular and Endovascular Surgery, Emory University School of Medicine, Atlanta, Georgia
| | | | - Salman Sher
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Stephanie Clement
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Khuram Ashraf
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Amr Rashed
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Tarek Kabbany
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Robert Neuman
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Alanna Morris
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Arshad Ali
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Salim Hayek
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - John Oshinski
- Department of Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Young-sup Yoon
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Edmund K Waller
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia7Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
210
|
Liao SY, Tse HF. Multipotent (adult) and pluripotent stem cells for heart regeneration: what are the pros and cons? Stem Cell Res Ther 2013; 4:151. [PMID: 24476362 PMCID: PMC4056686 DOI: 10.1186/scrt381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heart failure after myocardial infarction is the leading cause of mortality and morbidity worldwide. Existing medical and interventional therapies can only reduce the loss of cardiomyocytes during myocardial infarction but are unable to replenish the permanent loss of cardiomyocytes after the insult, which contributes to progressive pathological left ventricular remodeling and progressive heart failure. As a result, cell-based therapies using multipotent (adult) stem cells and pluripotent stem cells (embryonic stem cells or induced pluripotent stem cells) have been explored as potential therapeutic approaches to restore cardiac function in heart failure. Nevertheless, the optimal cell type with the best therapeutic efficacy and safety for heart regeneration is still unknown. In this review, the potential pros and cons of different types of multipotent (adult) stem cells and pluripotent stem cells that have been investigated in preclinical and clinical studies are reviewed, and the future perspective of stem cell-based therapy for heart regeneration is discussed.
Collapse
|
211
|
Kang YJ, Zheng L. Rejuvenation: an integrated approach to regenerative medicine. Regen Med Res 2013; 1:7. [PMID: 25984326 PMCID: PMC4376090 DOI: 10.1186/2050-490x-1-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 10/02/2013] [Indexed: 02/05/2023] Open
Abstract
The word "rejuvenate" found in the Merriam-Webster dictionary is (1) to make young or youthful again: give new vigor to, and (2) to restore to an original or new state. Regenerative medicine is the process of creating living, functional tissues to repair or replace tissue or organ function lost due to age, disease, damage, or congenital defects. To accomplish this, approaches including transplantation, tissue engineering, cell therapy, and gene therapy are brought into action. These all use exogenously prepared materials to forcefully mend the failed organ. The adaptation of the materials in the host and their integration into the organ are all uncertain. It is a common sense that tissue injury in the younger is easily repaired and the acute injury is healed better and faster. Why does the elder have a diminished capacity of self-repairing, or why does chronic injury cause the loss of the self-repairing capacity? There must be some critical elements that are involved in the repair process, but are suppressed in the elder or under the chronic injury condition. Rejuvenation of the self-repair mechanism would be an ideal solution for functional recovery of the failed organ. To achieve this, it would involve renewal of the injury signaling, reestablishment of the communication and transportation system, recruitment of the materials for repairing, regeneration of the failed organ, and rehabilitation of the renewed organ. It thus would require a comprehensive understanding of developmental biology and a development of new approaches to activate the critical players to rejuvenate the self-repair mechanism in the elder or under chronic injury condition. Efforts focusing on rejuvenation would expect an alternative, if not a better, accomplishment in the regenerative medicine.
Collapse
Affiliation(s)
- Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041 P.R. China ; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292 USA
| | - Lily Zheng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041 P.R. China
| |
Collapse
|
212
|
Doppler SA, Deutsch MA, Lange R, Krane M. Cardiac regeneration: current therapies-future concepts. J Thorac Dis 2013; 5:683-97. [PMID: 24255783 DOI: 10.3978/j.issn.2072-1439.2013.08.71] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease (CVD) continues to be one of the main causes of death in the western world. A high burden of disease and the high costs for the healthcare systems claim for novel therapeutic strategies besides current conventional medical care. One decade ago first clinical trials addressed stem cell based therapies as a potential alternative therapeutic strategy for myocardial regeneration and repair. Besides bone marrow derived stem cells (BMCs), adult stem cells from adipose or cardiac tissue have been used in current clinical studies with inconsistent results. Although outcomes in terms of safety and feasibility are generally encouraging, functional improvements were mostly disappointingly low and have failed to reach expectations. In the future, new concepts for myocardial regeneration, especially concerning recovery of cardiomyocyte loss, have to be developed. Transplantation of novel stem or progenitor cell populations with "true" regenerative potential, direct reprogramming of scar tissue into functional myocardium, tissue engineering or stimulation of endogenous cardiac repair by pharmacological agents are conceivable. This review summarizes current evidence of stem cell based regenerative therapies and discusses future strategies to improve functional outcomes.
Collapse
Affiliation(s)
- Stefanie A Doppler
- Department of Experimental Surgery, Department of Cardiovascular Surgery, German Heart Center Munich, Technische Universität München (TUM), Munich Heart Alliance, Munich, Germany
| | | | | | | |
Collapse
|
213
|
Mao J, Lv Z, Zhuang Y. MicroRNA-23a is involved in tumor necrosis factor-α induced apoptosis in mesenchymal stem cells and myocardial infarction. Exp Mol Pathol 2013; 97:23-30. [PMID: 24269648 DOI: 10.1016/j.yexmp.2013.11.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 11/01/2013] [Accepted: 11/12/2013] [Indexed: 01/16/2023]
Abstract
Cell therapy has emerged as an attractive therapeutic modality to treat myocardial infarction (MI) via repairing damaged myocardium, and mesenchymal stem cells (MSCs) are an appealing therapeutic approach for cardiac regeneration. However, the clinical application of MSC-based therapy is restricted because of the poor survival of implanted cells, and this poor survival remains poorly understood. Using a tumor necrosis factor (TNF)-α-induced bone marrow (BM)-MSC injury model in vitro and a rat MI model in vivo, we showed in the current study that miR-23a was involved in TNF-α-induced BM-MSC apoptosis through regulating caspase-7 and that the injection of BM-MSCs overexpressing miR-23a could improve left ventricular (LV) function and reduce infarct size in the rat MI model. Our findings elucidate the etiology of MI and provide an alternative treatment strategy for patients with heart failure caused by MI who are not optimal candidates for surgical treatment.
Collapse
Affiliation(s)
- Jianqiang Mao
- Department of Cardiovascular Surgery, Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China; Department of Cardiovascular Surgery, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China
| | - Zhiqian Lv
- Department of Cardiovascular Surgery, Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.
| | - Yu Zhuang
- Department of Cardiovascular Surgery, Sixth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China
| |
Collapse
|
214
|
Developing stem cell therapeutics for the heart also requires targeting non-myocytes. Heart Lung Circ 2013; 22:975-9. [PMID: 24231432 DOI: 10.1016/j.hlc.2013.10.091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
215
|
Abstract
Dilated cardiomyopathy is a serious and life-threatening disorder in children. It is the most common form of pediatric cardiomyopathy. Therapy for this condition has varied little over the last several decades and mortality continues to be high. Currently, children with dilated cardiomyopathy are treated with pharmacological agents and mechanical support, but most require heart transplantation and survival rates are not optimal. The lack of common treatment guidelines and inadequate survival rates after transplantation necessitates more therapeutic clinical trials. Stem cell and cell-based therapies offer an innovative approach to restore cardiac structure and function towards normal, possibly reducing the need for aggressive therapies and cardiac transplantation. Mesenchymal stem cells and cardiac stem cells may be the most promising cell types for treating children with dilated cardiomyopathy. The medical community must begin a systematic investigation of the benefits of current and novel treatments such as stem cell therapies for treating pediatric dilated cardiomyopathy.
Collapse
Affiliation(s)
- Sarah M Selem
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building/Room 908, PO Box 016960 (R-125), Miami, FL 33101, USA
| | | | | |
Collapse
|
216
|
Abstract
Vasculogenesis and angiogenesis are the major forms of blood vessel formation. Angiogenesis is the process where new vessels grow from pre-existing blood vessels, and is very important in the functional recovery of pathological conditions, such as wound healing and ischemic heart diseases. The development of better animal model and imaging technologies in past decades has greatly enriched our understanding on vasculogenesis and angiogenesis processes. Hypoxia turned out to be an important driving force for angiogenesis in various ischemic conditions. It stimulates expression of many growth factors like vascular endothelial growth factor, platelet-derived growth factor, insulin-like growth factor, and fibroblast growth factor, which play critical role in induction of angiogenesis. Other cellular components like monocytes, T cells, neutrophils, and platelets also play significant role in induction and regulation of angiogenesis. Various stem/progenitor cells also being recruited to the ischemic sites play crucial role in the angiogenesis process. Pre-clinical studies showed that stem/progenitor cells with/without combination of growth factors induce neovascularization in the ischemic tissues in various animal models. In this review, we will discuss about the fundamental factors that regulate the angiogenesis process and the use of stem cells as therapeutic regime for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Jingwei Lu
- Cardiovascular Stem Cell Research Laboratory, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, 460W, 12th Avenue, BRT 382, Columbus, OH, 43210, USA
| | | | | |
Collapse
|
217
|
Park KE, Moyé LA, Henry TD, Perin EC, Sayre SL, Bettencourt J, Vojvodic RW, Olson RE, Pepine CJ. Implementation of cardovascular cell therapy network trials: challenges, innovation and lessons learned from experience in the CCTRN. Expert Rev Cardiovasc Ther 2013; 11:1495-502. [PMID: 24147517 DOI: 10.1586/14779072.2013.839943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The cardiovascular cell therapy network was developed by the National Heart, Lung and Blood Institute to design and conduct clinical trials to advance the field of cardiovascular (CV) cell-based therapy. The Cardiovascular Cell Therapy Network successfully completed three clinical trials involving approximately 300 subjects across five centers and six satellites. Although the concept of a network within clinical trials research is not new, the knowledge gained in the implementation of such large-scale trials, particularly in novel therapeutic areas such as cell therapy is not often detailed in the literature. The purpose of this communication is to summarize key factors in achieving network goals and share the knowledge gained to promote success in future cardiovascular disease cell therapy trials and networks.
Collapse
Affiliation(s)
- Ki E Park
- University of Florida College of Medicine, 1600 SW Archer Rd. Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Jang MY, Chun SI, Mun CW, Hong KS, Shin JW. Evaluation of metabolomic changes as a biomarker of chondrogenic differentiation in 3D-cultured human mesenchymal stem cells using proton (1H) nuclear magnetic resonance spectroscopy. PLoS One 2013; 8:e78325. [PMID: 24205199 PMCID: PMC3804484 DOI: 10.1371/journal.pone.0078325] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 09/11/2013] [Indexed: 01/09/2023] Open
Abstract
PURPOSE The purpose of this study was to evaluate the metabolomic changes in 3D-cultured human mesenchymal stem cells (hMSCs) in alginate beads, so as to identify biomarkers during chondrogenesis using (1)H nuclear magnetic resonance (NMR) spectroscopy. MATERIALS AND METHODS hMSCs (2×10(6) cells/mL) were seeded into alginate beads, and chondrogenesis was allowed to progress for 15 days. NMR spectra of the chondrogenic hMSCs were obtained at 4, 7, 11, and 15 days using a 14.1-T (600-MHz) NMR with the water suppression sequence, zgpr. Real-Time polymerase chain reaction (PCR) was performed to confirm that that the hMSCs differentiated into chondrocytes and to analyze the metabolomic changes indicated by the NMR spectra. RESULTS During chondrogenesis, changes were detected in several metabolomes as hMSC chondrogenesis biomarkers, e.g., fatty acids, alanine, glutamate, and phosphocholine. The metabolomic changes were compared with the Real-Time PCR results, and significant differences were determined using statistical analysis. We found that changes in metabolomes were closely related to biological reactions that occurred during the chondrogenesis of hMSCs. CONCLUSIONS In this study, we confirm that metabolomic changes detected by (1)H-NMR spectroscopy during chondrogenic differentiation of 3D-cultured hMSCs in alginate beads can be considered as biomarkers of stem cell differentiation.
Collapse
Affiliation(s)
- Moo-Young Jang
- Department of Biomedical Engineering/UHRC, Inje University, Gimhae, Gyeongnam, South Korea
| | - Song-I Chun
- Department of Biomedical Engineering/UHRC, Inje University, Gimhae, Gyeongnam, South Korea
| | - Chi-Woong Mun
- Department of Biomedical Engineering/UHRC, Inje University, Gimhae, Gyeongnam, South Korea
- Graduate School of Health Science and Technology, Inje University, Gimhae, Gyeongnam, South Korea
| | - Kwan Soo Hong
- Korea Basic Science Institute, Ochang, Chungbuk, South Korea
| | - Jung-Woog Shin
- Department of Biomedical Engineering/UHRC, Inje University, Gimhae, Gyeongnam, South Korea
- Graduate School of Health Science and Technology, Inje University, Gimhae, Gyeongnam, South Korea
| |
Collapse
|
219
|
Du Y, Lai PT, Leung CH, Pong PWT. Design of superparamagnetic nanoparticles for magnetic particle imaging (MPI). Int J Mol Sci 2013; 14:18682-710. [PMID: 24030719 PMCID: PMC3794803 DOI: 10.3390/ijms140918682] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 07/29/2013] [Accepted: 08/14/2013] [Indexed: 01/21/2023] Open
Abstract
Magnetic particle imaging (MPI) is a promising medical imaging technique producing quantitative images of the distribution of tracer materials (superparamagnetic nanoparticles) without interference from the anatomical background of the imaging objects (either phantoms or lab animals). Theoretically, the MPI platform can image with relatively high temporal and spatial resolution and sensitivity. In practice, the quality of the MPI images hinges on both the applied magnetic field and the properties of the tracer nanoparticles. Langevin theory can model the performance of superparamagnetic nanoparticles and predict the crucial influence of nanoparticle core size on the MPI signal. In addition, the core size distribution, anisotropy of the magnetic core and surface modification of the superparamagnetic nanoparticles also determine the spatial resolution and sensitivity of the MPI images. As a result, through rational design of superparamagnetic nanoparticles, the performance of MPI could be effectively optimized. In this review, the performance of superparamagnetic nanoparticles in MPI is investigated. Rational synthesis and modification of superparamagnetic nanoparticles are discussed and summarized. The potential medical application areas for MPI, including cardiovascular system, oncology, stem cell tracking and immune related imaging are also analyzed and forecasted.
Collapse
Affiliation(s)
- Yimeng Du
- Department of Electrical and Electronic Engineering, the University of Hong Kong, Hong Kong; E-Mails: (Y.D.); (P.T.L.); (C.H.L.)
| | - Pui To Lai
- Department of Electrical and Electronic Engineering, the University of Hong Kong, Hong Kong; E-Mails: (Y.D.); (P.T.L.); (C.H.L.)
| | - Cheung Hoi Leung
- Department of Electrical and Electronic Engineering, the University of Hong Kong, Hong Kong; E-Mails: (Y.D.); (P.T.L.); (C.H.L.)
| | - Philip W. T. Pong
- Department of Electrical and Electronic Engineering, the University of Hong Kong, Hong Kong; E-Mails: (Y.D.); (P.T.L.); (C.H.L.)
| |
Collapse
|
220
|
Sreejit P, Verma RS. Natural ECM as biomaterial for scaffold based cardiac regeneration using adult bone marrow derived stem cells. Stem Cell Rev Rep 2013; 9:158-71. [PMID: 23319217 DOI: 10.1007/s12015-013-9427-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cellular therapy using stem cells for cardiac diseases has recently gained much interest in the scientific community due to its potential in regenerating damaged and even dead tissue and thereby restoring the organ function. Stem cells from various sources and origin are being currently used for regeneration studies directly or along with differentiation inducing agents. Long term survival and minimal side effects can be attained by using autologous cells and reduced use of inducing agents. Cardiomyogenic differentiation of adult derived stem cells has been previously reported using various inducing agents but the use of a potentially harmful DNA demethylating agent 5-azacytidine (5-azaC) has been found to be critical in almost all studies. Alternate inducing factors and conditions/stimulant like physical condition including electrical stimulation, chemical inducers and biological agents have been attempted by numerous groups to induce cardiac differentiation. Biomaterials were initially used as artificial scaffold in in vitro studies and later as a delivery vehicle. Natural ECM is the ideal biological scaffold since it contains all the components of the tissue from which it was derived except for the living cells. Constructive remodeling can be performed using such natural ECM scaffolds and stem cells since, the cells can be delivered to the site of infraction and once delivered the cells adhere and are not "lost". Due to the niche like conditions of ECM, stem cells tend to differentiate into tissue specific cells and attain several characteristics similar to that of functional cells even in absence of any directed differentiation using external inducers. The development of niche mimicking biomaterials and hybrid biomaterial can further advance directed differentiation without specific induction. The mechanical and electrical integration of these materials to the functional tissue is a problem to be addressed. The search for the perfect extracellular matrix for therapeutic applications including engineering cardiac tissue structures for post ischemic cardiac tissue regeneration continues.
Collapse
Affiliation(s)
- P Sreejit
- Stem Cell and Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, TN, India
| | | |
Collapse
|
221
|
Allukian M, Xu J, Morris M, Caskey R, Dorsett-Martin W, Plappert T, Griswold M, Gorman JH, Gorman RC, Liechty KW. Mammalian cardiac regeneration after fetal myocardial infarction requires cardiac progenitor cell recruitment. Ann Thorac Surg 2013; 96:163-70. [PMID: 23816072 DOI: 10.1016/j.athoracsur.2013.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 04/01/2013] [Accepted: 04/02/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND In contrast to the adult, fetal sheep consistently regenerate functional myocardium after myocardial infarction. We hypothesize that this regeneration is due to the recruitment of cardiac progenitor cells to the infarct by stromal-derived factor-1α (SDF-1α) and that its competitive inhibition will block the regenerative fetal response. METHODS A 20% apical infarct was created in adult and fetal sheep by selective permanent coronary artery ligation. Lentiviral overexpression of mutant SDF-1α competitively inhibited SDF-1α in fetal infarcts. Echocardiography was performed to assess left ventricular function and infarct size. Cardiac progenitor cell recruitment and proliferation was assessed in fetal infarcts at 1 month by immunohistochemistry for nkx2.5 and 5-bromo-2-deoxyuridine. RESULTS Competitive inhibition of SDF-1α converted the regenerative fetal response into a reparative response, similar to the adult. SDF-inhibited fetal infarcts demonstrated significant infarct expansion by echocardiography (p < 0.001) and a significant decrease in the number of nkx2.5+ cells repopulating the infarct (p < 0.001). CONCLUSIONS The fetal regenerative response to myocardial infarction requires the recruitment of cardiac progenitor cells and is dependent on SDF1α. This novel model of mammalian cardiac regeneration after myocardial infarction provides a powerful tool to better understand cardiac progenitor cell biology and to develop strategies to cardiac regeneration in the adult.
Collapse
Affiliation(s)
- Myron Allukian
- Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Bernstein HS, Samad T, Cholsiripunlert S, Khalifian S, Gong W, Ritner C, Aurigui J, Ling V, Wilschut KJ, Bennett S, Hoffman J, Oishi P. Stem cell antigen-1 in skeletal muscle function. PLOS CURRENTS 2013; 5. [PMID: 24042315 PMCID: PMC3770837 DOI: 10.1371/currents.md.411a8332d61e22725e6937b97e6d0ef8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stem cell antigen-1 (Sca-1) is a member of the Ly-6 multigene family encoding highly homologous, glycosyl-phosphatidylinositol-anchored membrane proteins. Sca-1 is expressed on muscle-derived stem cells and myogenic precursors recruited to sites of muscle injury. We previously reported that inhibition of Sca-1 expression stimulated myoblast proliferation in vitro and regulated the tempo of muscle repair in vivo. Despite its function in myoblast expansion during muscle repair, a role for Sca-1 in normal, post-natal muscle has not been thoroughly investigated. We systematically compared Sca-1-/- (KO) and Sca-1+/+ (WT) mice and hindlimb muscles to elucidate the tissue, contractile, and functional effects of Sca-1 in young and aging animals. Comparison of muscle volume, fibrosis, myofiber cross-sectional area, and Pax7+ myoblast number showed little differences between ages or genotypes. Exercise protocols, however, demonstrated decreased stamina in KO versus WT mice, with young KO mice achieving results similar to aging WT animals. In addition, KO mice did not improve with practice, while WT animals demonstrated conditioning over time. Surprisingly, myomechanical analysis of isolated muscles showed that KO young muscle generated more force and experienced less fatigue. However, KO muscle also demonstrated incomplete relaxation with fatigue. These findings suggest that Sca-1 is necessary for muscle conditioning with exercise, and that deficient conditioning in Sca-1 KO animals becomes more pronounced with age.
Collapse
|
223
|
Abstract
Heart failure is a devastating condition, the progression of which culminates in a mismatch of oxygen supply and demand, with limited options for treatment. Heart failure has several underlying causes including, but not limited to, ischaemic heart disease, valvular dysfunction, and hypertensive heart disease. Dysfunctional blood vessel formation is a major problem in advanced heart failure, regardless of the aetiology. Vascular endothelial growth factor (VEGF) is the cornerstone cytokine involved in the formation of new vessels. A multitude of investigations, at both the preclinical and clinical levels, have garnered valuable information on the potential utility of targeting VEGF as a treatment option for heart failure. However, clinical trials of VEGF gene therapy in patients with coronary artery disease or peripheral artery disease have not, to date, demonstrated clinical benefit. In this Review, we outline the biological characterization of VEGF, and examine the evidence for its potential therapeutic application, including the novel concept of VEGF as adjuvant therapy to stem cell transplantation, in patients with heart failure.
Collapse
|
224
|
Li Y, Guo X, Xue Q, Zhu M, Gao L, Wang Y. Single cell gene profiling revealed heterogeneity of paracrine effects of bone marrow cells in mouse infarcted hearts. PLoS One 2013; 8:e68270. [PMID: 23861876 PMCID: PMC3702556 DOI: 10.1371/journal.pone.0068270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 05/28/2013] [Indexed: 12/25/2022] Open
Abstract
It is now recognized that transplantation of bone marrow cells (BMCs) into infarcted hearts has the capacity to improve the cardiac function through paracrine effects. However, detailed expression levels of paracrine factors in BMCs in infarcted hearts are poorly described. By use of laser capture microdissection combined with real-time PCR, we depicted the expression profiles of paracrine factors in infarcted hearts versus normal hearts. Consistent with the in vivo observation, a similar expression pattern was evidenced in cultured BMCs. Furthermore, BMCs displayed heterogeneity of paracrine effects in infarcted hearts as analyzed at the single cell level using single cell PCR. Interestingly, the CD45+ subpopulation showed higher expression levels of angiogenic factors compared to other subpopulations. Finally, most angiogenic factors were induced under the microenvironment of infarction. Our study demonstrated the heterogeneity of paracrine effects in BMCs at single cell level in infarcted hearts, highlighting preferential expression of angiogenic factors in the CD45+ subpopulation. These findings broaden our understanding of paracrine effects of BMCs in vivo, and offer new insights into BMCs therapy in myocardial infarction (MI).
Collapse
Affiliation(s)
- Yanhua Li
- Institute of Geriatric Cardiology, the General Hospital of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Xinhong Guo
- Institute of Geriatric Cardiology, the General Hospital of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Qiao Xue
- Institute of Geriatric Cardiology, the General Hospital of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Mei Zhu
- Institute of Geriatric Cardiology, the General Hospital of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Lei Gao
- Institute of Geriatric Cardiology, the General Hospital of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Yu Wang
- Institute of Geriatric Cardiology, the General Hospital of Chinese People’s Liberation Army (PLA), Beijing, China
- * E-mail:
| |
Collapse
|
225
|
Kim WS, Lee S, Yoon YS. Cardiovascular repair with bone marrow-derived cells. Blood Res 2013; 48:76-86. [PMID: 23826576 PMCID: PMC3698412 DOI: 10.5045/br.2013.48.2.76] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 12/31/2022] Open
Abstract
While bone marrow (BM)-derived cells have been comprehensively studied for their propitious pre-clinical results, clinical trials have shown controversial outcomes. Unlike previously acknowledged, more recent studies have now confirmed that humoral and paracrine effects are the key mechanisms for tissue regeneration and functional recovery, instead of transdifferentiation of BM-derived cells into cardiovascular tissues. The progression of the understanding of BM-derived cells has further led to exploring efficient methods to isolate and obtain, without mobilization, sufficient number of cell populations that would eventually have a higher therapeutic potential. As such, hematopoietic CD31+ cells, prevalent in both bone marrow and peripheral blood, have been discovered, in recent studies, to have angiogenic and vasculogenic activities and to show strong potential for therapeutic neovascularization in ischemic tissues. This article will discuss recent advancement on BM-derived cell therapy and the implication of newly discovered CD31+ cells.
Collapse
Affiliation(s)
- Woan-Sang Kim
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, GA, USA
| | | | | |
Collapse
|
226
|
Chow K, Fessel JP, Kaoriihida-Stansbury, Schmidt EP, Gaskill C, Alvarez D, Graham B, Harrison DG, Wagner DH, Nozik-Grayck E, West JD, Klemm DJ, Majka SM. Dysfunctional resident lung mesenchymal stem cells contribute to pulmonary microvascular remodeling. Pulm Circ 2013; 3:31-49. [PMID: 23662173 PMCID: PMC3641738 DOI: 10.4103/2045-8932.109912] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary vascular remodeling and oxidative stress are common to many adult lung diseases. However, little is known about the relevance of lung mesenchymal stem cells (MSCs) in these processes. We tested the hypothesis that dysfunctional lung MSCs directly participate in remodeling of the microcirculation. We employed a genetic model to deplete extracellular superoxide dismutase (EC-SOD) in lung MSCs coupled with lineage tracing analysis. We crossed floxpsod3 and mT/mG reporter mice to a strain expressing Cre recombinase under the control of the ABCG2 promoter. We demonstrated In vivo that depletion of EC-SOD in lung MSCs resulted in their contribution to microvascular remodeling in the smooth muscle actin positive layer. We further characterized lung MSCs to be multipotent vascular precursors, capable of myofibroblast, endothelial and pericyte differentiation in vitro. EC-SOD deficiency in cultured lung MSCs accelerated proliferation and apoptosis, restricted colony-forming ability, multilineage differentiation potential and promoted the transition to a contractile phenotype. Further studies correlated cell dysfunction to alterations in canonical Wnt/β-catenin signaling, which were more evident under conditions of oxidative stress. Our data establish that lung MSCs are a multipotent vascular precursor population, a population which has the capacity to participate in vascular remodeling and their function is likely regulated in part by the Wnt/β-catenin signaling pathway. These studies highlight an important role for microenviromental regulation of multipotent MSC function as well as their potential to contribute to tissue remodeling.
Collapse
Affiliation(s)
- Kelsey Chow
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Fukata M, Ishikawa F, Najima Y, Yamauchi T, Saito Y, Takenaka K, Miyawaki K, Shimazu H, Shimoda K, Kanemaru T, Nakamura KI, Odashiro K, Nagafuji K, Harada M, Akashi K. Contribution of bone marrow-derived hematopoietic stem/progenitor cells to the generation of donor-marker⁺ cardiomyocytes in vivo. PLoS One 2013; 8:e62506. [PMID: 23667482 PMCID: PMC3647070 DOI: 10.1371/journal.pone.0062506] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 03/26/2013] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Definite identification of the cell types and the mechanism relevant to cardiomyogenesis is essential for effective cardiac regenerative medicine. We aimed to identify the cell populations that can generate cardiomyocytes and to clarify whether generation of donor-marker(+) cardiomyocytes requires cell fusion between BM-derived cells and recipient cardiomyocytes. METHODOLOGY/PRINCIPAL FINDINGS Purified BM stem/progenitor cells from green fluorescence protein (GFP) mice were transplanted into C57BL/6 mice or cyan fluorescence protein (CFP)-transgenic mice. Purified human hematopoietic stem cells (HSCs) from cord blood were transplanted into immune-compromised NOD/SCID/IL2rγ(null) mice. GFP(+) cells in the cardiac tissue were analyzed for the antigenecity of a cardiomyocyte by confocal microscopy following immunofluorescence staining. GFP(+) donor-derived cells, GFP(+)CFP(+) fused cells, and CFP(+) recipient-derived cells were distinguished by linear unmixing analysis. Hearts of xenogeneic recipients were evaluated for the expression of human cardiomyocyte genes by real-time quantitative polymerase chain reaction. In C57BL/6 recipients, Lin(-/low)CD45(+) hematopoietic cells generated greater number of GFP(+) cardiomyocytes than Lin(-/low)CD45(-) mesenchymal cells (37.0+/-23.9 vs 0.00+/-0.00 GFP(+) cardiomyocytes per a recipient, P = 0.0095). The number of transplanted purified HSCs (Lin(-/low)Sca-1(+) or Lin(-)Sca-1(+)c-Kit(+) or CD34(-)Lin(-)Sca-1(+)c-Kit(+)) showed correlation to the number of GFP(+) cardiomyocytes (P<0.05 in each cell fraction), and the incidence of GFP(+) cardiomyocytes per injected cell dose was greatest in CD34(-)Lin(-)Sca-1(+)c-Kit(+) recipients. Of the hematopoietic progenitors, total myeloid progenitors generated greater number of GFP(+) cardiomyocytes than common lymphoid progenitors (12.8+/-10.7 vs 0.67+/-1.00 GFP(+) cardiomyocytes per a recipient, P = 0.0021). In CFP recipients, all GFP(+) cardiomyocytes examined coexpressed CFP. Human troponin C and myosin heavy chain 6 transcripts were detected in the cardiac tissue of some of the xenogeneic recipients. CONCLUSIONS/SIGNIFICANCE Our results indicate that HSCs resulted in the generation of cardiomyocytes via myeloid intermediates by fusion-dependent mechanism. The use of myeloid derivatives as donor cells could potentially allow more effective cell-based therapy for cardiac repair.
Collapse
Affiliation(s)
- Mitsuhiro Fukata
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
- * E-mail: (MF); (FI)
| | - Fumihiko Ishikawa
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
- Laboratory for Human Disease Models, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
- * E-mail: (MF); (FI)
| | - Yuho Najima
- Laboratory for Human Disease Models, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Yoriko Saito
- Laboratory for Human Disease Models, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
| | - Katsuto Takenaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Hideki Shimazu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Kazuya Shimoda
- Department of Gastroenterology and Hematology, Faculty of Medicine, Miyazaki University, Miyazaki, Japan
| | | | - Kei-ichiro Nakamura
- Second Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Keita Odashiro
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Koji Nagafuji
- Division of Hematology and Oncology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Mine Harada
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
228
|
Shaharuddin B, Ahmad S, Ali S, Meeson A. Limbal side population cells: a future treatment for limbal stem cell deficiency. Regen Med 2013; 8:319-31. [DOI: 10.2217/rme.13.18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Corneal blindness carries a morbidity that affects quality of life and is often associated with an increased economic burden. In this review, we focus on the severe and painful condition of limbal stem cell deficiency, an important cause of corneal blindness. Conventional corneal transplantation usually results in graft failure and is contraindicated in this condition. Ex vivo-expanded limbal epithelial transplantation has been used as a cellular-based therapy to regenerate and reconstruct the ocular surface as a mode of treatment. Enrichment methods for stem cells are a strategy to improve the outcome of limbal stem cell transplantation. Here we discuss the side population assay as a functional assay to enrich for stem cells as an important source of limbal stem cells. The challenges in ex vivo-expanded limbal stem cell transplantation are wide and varied and will be addressed in this review with regard to improving the clinical outcomes of cultivated limbal stem cell transplantation.
Collapse
Affiliation(s)
- Bakiah Shaharuddin
- Advanced Medical & Dental Institute, Universiti Sains Malaysia, 13200 Pulau Pinang, Malaysia
| | - Sajjad Ahmad
- St Paul’s Eye Unit, Royal Liverpool University Hospital, Prescot Street, Liverpool, L7 8XP, UK
- Department of Eye & Vision Sciences, Institute of Ageing & Chronic Disease, University of Liverpool, 4th Floor UCD Building, Daulby Street, Liverpool, L69 3GA, UK
| | - Simi Ali
- Institute of Cellular Medicine, William-Leech Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Annette Meeson
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
229
|
Schoenfeld M, Frishman WH, Leri A, Kajstura J, Anversa P. The existence of myocardial repair: mechanistic insights and enhancements. Cardiol Rev 2013; 21:111-20. [PMID: 23568056 PMCID: PMC3946988 DOI: 10.1097/crd.0b013e318289d7a9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The lack of myocardial repair after myocardial infarction and the heart failure that eventually ensues was thought of as proof that myocardial cell regeneration and myocardial repair mechanisms do not exist. Recently, growing experimental and clinical evidence has proven this concept wrong. Cardiac stem cells and endogenous myocardial repair mechanisms do exist; however, they do not produce significant myocardial repair. Similarly, the preliminary results of stem cell therapy for myocardial repair have shown early promise but modest results. Preclinical studies are the key to understanding stem cell senescence and lack of cellular contact and vasculature in the infarcted region. Additional laboratory studies are sure to unlock the therapeutic mechanisms that will be required for significant myocardial repair.
Collapse
Affiliation(s)
- Matthew Schoenfeld
- Department of Medicine, New York Presbyterian/Weill Cornell Medical Center, New York, NY
| | - William H. Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| | - Annarosa Leri
- Departments of Anesthesia and Medicine, Harvard Medical School/Brigham & Womens Hospital, Boston, MA
| | - Jan Kajstura
- Departments of Anesthesia and Medicine, Harvard Medical School/Brigham & Womens Hospital, Boston, MA
| | - Piero Anversa
- Departments of Anesthesia and Medicine, Harvard Medical School/Brigham & Womens Hospital, Boston, MA
| |
Collapse
|
230
|
Wang M, Yu Q, Wang L, Gu H. Distinct patterns of histone modifications at cardiac-specific gene promoters between cardiac stem cells and mesenchymal stem cells. Am J Physiol Cell Physiol 2013; 304:C1080-90. [PMID: 23552285 DOI: 10.1152/ajpcell.00359.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) and cardiac stem cells (CSCs) possess different potential to develop into cardiomyocytes. The mechanism underlying cardiomyogenic capacity of MSCs and CSCs remains elusive. It is well established that histone modifications correlate with gene expression and contribute to cell fate commitment. Here we hypothesize that specific histone modifications accompany cardiac-specific gene expression, thus determining the differentiation capacity of MSCs and CSCs toward heart cells. Our results indicate that, at the promoter regions of cardiac-specific genes (Myh6, Myl2, Actc1, Tnni3, and Tnnt2), the levels of histone acetylation of H3 (acH3) and H4 (acH4), as a mark indicative of gene activation, were higher in CSCs (Sca-1(+)CD29(+)) than MSCs. Additionally, lower binding levels of histone deacetylase (HDAC) 1 and HDAC2 at promoter regions of cardiac-specific genes were noticed in CSCs than MSCs. Treatment with trichostatin A, an HDAC inhibitor, upregulated cardiac-specific gene expression in MSCs. Suppression of HDAC1 or HDAC2 expression by small interfering RNAs led to increased cardiac gene expression and was accompanied by enhanced acH3 and acH4 levels at gene loci. We conclude that greater levels of histone acetylation at cardiac-specific gene loci in CSCs than MSCs reflect a stronger potential for CSCs to develop into cardiomyocytes. These lineage-differential histone modifications are likely due to less HDAC recruitment at cardiac-specific gene promoters in CSCs than MSCs.
Collapse
Affiliation(s)
- Meijing Wang
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | | | | | |
Collapse
|
231
|
Petriz J. Flow cytometry of the side population (SP). CURRENT PROTOCOLS IN CYTOMETRY 2013; Chapter 9:9.23.1-9.23.20. [PMID: 23546779 DOI: 10.1002/0471142956.cy0923s64] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The side population (SP) has become an important hallmark for the definition of the stem-cell compartment, especially for the detection of stem cells and for their physical isolation by fluorescence-activated cell sorting (FACS). SP cells are CD34(-) and were discovered using ultraviolet excitation based on the efflux of Hoechst 33342 (Ho342). Although the method works as originally described, the protocol is difficult for most investigators to perform: first, because the ability to discriminate SP cells is based on the differential retention of Ho342 during a functional assay; second, because of the difficulties in setting the right experimental and acquisition conditions; and third, because analysis of the acquired data requires extensive expertise in flow cytometry to accurately detect the SP events. More recently, a new assay based on the efflux of Vybrant DyeCycle Violet stain (DCV) has been documented to discriminate SP cells. This unit contains many helpful pointers to aid the user in obtaining the best possible results with these assays.
Collapse
Affiliation(s)
- Jordi Petriz
- Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
232
|
Saritas EU, Goodwill PW, Croft LR, Konkle JJ, Lu K, Zheng B, Conolly SM. Magnetic particle imaging (MPI) for NMR and MRI researchers. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2013; 229:116-26. [PMID: 23305842 PMCID: PMC3602323 DOI: 10.1016/j.jmr.2012.11.029] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/21/2012] [Accepted: 11/30/2012] [Indexed: 05/04/2023]
Abstract
Magnetic Particle Imaging (MPI) is a new tracer imaging modality that is gaining significant interest from NMR and MRI researchers. While the physics of MPI differ substantially from MRI, it employs hardware and imaging concepts that are familiar to MRI researchers, such as magnetic excitation and detection, pulse sequences, and relaxation effects. Furthermore, MPI employs the same superparamagnetic iron oxide (SPIO) contrast agents that are sometimes used for MR angiography and are often used for MRI cell tracking studies. These SPIOs are much safer for humans than iodine or gadolinium, especially for Chronic Kidney Disease (CKD) patients. The weak kidneys of CKD patients cannot safely excrete iodine or gadolinium, leading to increased morbidity and mortality after iodinated X-ray or CT angiograms, or after gadolinium-MRA studies. Iron oxides, on the other hand, are processed in the liver, and have been shown to be safe even for CKD patients. Unlike the "black blood" contrast generated by SPIOs in MRI due to increased T2* dephasing, SPIOs in MPI generate positive, "bright blood" contrast. With this ideal contrast, even prototype MPI scanners can already achieve fast, high-sensitivity, and high-contrast angiograms with millimeter-scale resolutions in phantoms and in animals. Moreover, MPI shows great potential for an exciting array of applications, including stem cell tracking in vivo, first-pass contrast studies to diagnose or stage cancer, and inflammation imaging in vivo. So far, only a handful of prototype small-animal MPI scanners have been constructed worldwide. Hence, MPI is open to great advances, especially in hardware, pulse sequence, and nanoparticle improvements, with the potential to revolutionize the biomedical imaging field.
Collapse
Affiliation(s)
- Emine U Saritas
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762, USA.
| | | | | | | | | | | | | |
Collapse
|
233
|
Catacchio I, Berardi S, Reale A, De Luisi A, Racanelli V, Vacca A, Ria R. Evidence for bone marrow adult stem cell plasticity: properties, molecular mechanisms, negative aspects, and clinical applications of hematopoietic and mesenchymal stem cells transdifferentiation. Stem Cells Int 2013; 2013:589139. [PMID: 23606860 PMCID: PMC3625599 DOI: 10.1155/2013/589139] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/22/2013] [Indexed: 12/24/2022] Open
Abstract
In contrast to the pluripotent embryonic stem cells (ESCs) which are able to give rise to all cell types of the body, mammalian adult stem cells (ASCs) appeared to be more limited in their differentiation potential and to be committed to their tissue of origin. Recently, surprising new findings have contradicted central dogmas of commitment of ASCs by showing their plasticity to differentiate across tissue lineage boundaries, irrespective of classical germ layer designations. The present paper supports the plasticity of the bone marrow stem cells (BMSCs), bringing the most striking and the latest evidences of the transdifferentiation properties of the bone marrow hematopoietic and mesenchymal stem cells (BMHSCs, and BMMSCs), the two BM populations of ASCs better characterized. In addition, we report the possible mechanisms that may explain these events, outlining the clinical importance of these phenomena and the relative problems.
Collapse
Affiliation(s)
- Ivana Catacchio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Simona Berardi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Antonia Reale
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Annunziata De Luisi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine, University of Bari Medical School, Policlinico, Piazza Giulio Cesare 11, I-70124 Bari, Italy
| |
Collapse
|
234
|
Kamiura N, Hirahashi J, Matsuzaki Y, Idei M, Takase O, Fujita T, Takato T, Hishikawa K. Basic helix-loop-helix transcriptional factor MyoR regulates BMP-7 in acute kidney injury. Am J Physiol Renal Physiol 2013; 304:F1159-66. [PMID: 23515721 DOI: 10.1152/ajprenal.00510.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
MyoR was originally identified as a transcriptional repressor in embryonic skeletal muscle precursors, but its function in adult kidney has not been clarified. In this study, we tried to clarify the functional role of MyoR using MyoR(-/-) mice. Cisplatin induced a significantly higher degree of severe renal dysfunction, tubular injury, and mortality in MyoR(-/-) mice than in wild-type mice. The injection of cisplatin significantly increased the number of apoptotic cells in the kidney tissues of MyoR(-/-) mice, compared with that in wild-type mice. To clarify the mechanism of severe cisplatin-induced damage and apoptosis in MyoR(-/-) mice, we focused on the p53 signaling pathway and bone morphogenic protein-7 (BMP-7). Treatment with cisplatin significantly activated p53 signaling in cultured renal proximal tubular epithelial cells (RTECs) in both wild-type and MyoR(-/-) mice, but no significant difference between the groups was observed. The injection of cisplatin significantly increased the expression of BMP-7 in the kidney tissues of wild-type mice, but no increase was observed in the MyoR(-/-) mice. Treatment with cisplatin significantly increased the expression of BMP-7 in cultured RTECs from wild-type mice but not in those from MyoR(-/-) mice. Moreover, treatment with recombinant BMP-7 rescued the cisplatin-induced apoptosis in RTECs from MyoR(-/-) mice. Taken together, our results demonstrate a new protective role of MyoR in adult kidneys that acts through the regulation of BMP-7.
Collapse
Affiliation(s)
- Nozomu Kamiura
- Department of Advanced Nephrology and Regenerative Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Law S, Chaudhuri S. Mesenchymal stem cell and regenerative medicine: regeneration versus immunomodulatory challenges. AMERICAN JOURNAL OF STEM CELLS 2013; 2:22-38. [PMID: 23671814 PMCID: PMC3636724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 02/12/2013] [Indexed: 06/02/2023]
Abstract
Mesenchymal Stem cells (MSC) are now presented with the opportunities of multifunctional therapeutic approaches. Several reports are in support of their self-renewal, capacity for multipotent differentiation, and immunomodulatory properties. They are unique to contribute to the regeneration of mesenchymal tissues such as bone, cartilage, muscle, ligament, tendon, and adipose. In addition to promising trials in regenerative medicine, such as in the treatment of major bone defects and myocardial infarction, MSC has shown a therapeutic effect other than direct hematopoiesis support in hematopoietic reconstruction. MSCs are identified by the expression of many molecules including CD105 (SH2) and CD73(SH3/4) and are negative for the hematopoietic markers CD34, CD45, and CD14. Manufacturing of MSC for clinical trials is also an important aspect as their differentiation, homing and Immunomodulatory properties may differ. Their suppressive effects on immune cells, including T cells, B cells, NK cells and DC cells, suggest MSCs as a novel therapy for GVHD and other autoimmune disorders. Since the cells by themselves are non-immunogenic, tissue matching between MSC donor and recipient is not essential and, MSC may be the first cell type able to be used as an "off-the-shelf" therapeutic product. Following a successful transplantation, the migration of MSC to the site of injury refers to the involvement of chemokines and chemokine receptors of respective specificity. It has been demonstrated that cultured MSCs have the ability to engraft into healthy as well as injured tissue and can differentiate into several cell types in vivo, which facilitates MSC to be an ideal tool for regenerative therapy in different disease types. However, some observations have raised questions about the limitations for proper use of MSC considering some critical factors that warn regular clinical use.
Collapse
Affiliation(s)
- Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine108 C R Avenue, Kolkata-700073, India
| | - Samaresh Chaudhuri
- Guest Faculty Professor, Biotechnology, West Bengal University of TechnologyKolkata, India
| |
Collapse
|
236
|
Dey D, Han L, Bauer M, Sanada F, Oikonomopoulos A, Hosoda T, Unno K, De Almeida P, Leri A, Wu JC. Dissecting the molecular relationship among various cardiogenic progenitor cells. Circ Res 2013; 112:1253-62. [PMID: 23463815 DOI: 10.1161/circresaha.112.300779] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Multiple progenitors derived from the heart and bone marrow (BM) have been used for cardiac repair. Despite this, not much is known about the molecular identity and relationship among these progenitors. To develop a robust stem cell therapy for the heart, it is critical to understand the molecular identity of the multiple cardiogenic progenitor cells. OBJECTIVE This study is the first report of high-throughput transcriptional profiling of cardiogenic progenitor cells carried out on an identical platform. METHOD AND RESULTS Microarray-based transcriptional profiling was carried out for 3 cardiac (ckit(+), Sca1(+), and side population) and 2 BM (ckit(+) and mesenchymal stem cell) progenitors, obtained from age- and sex-matched wild-type C57BL/6 mice. Analysis indicated that cardiac-derived ckit(+) population was very distinct from Sca1(+) and side population cells in the downregulation of genes encoding for cell-cell and cell-matrix adhesion proteins, and in the upregulation of developmental genes. Significant enrichment of transcripts involved in DNA replication and repair was observed in BM-derived progenitors. The BM ckit(+) cells seemed to have the least correlation with the other progenitors, with enrichment of immature neutrophil-specific molecules. CONCLUSIONS Our study indicates that cardiac ckit(+) cells represent the most primitive population in the rodent heart. Primitive cells of cardiac versus BM origin differ significantly with respect to stemness and cardiac lineage-specific genes, and molecules involved in DNA replication and repair. The detailed molecular profile of progenitors reported here will serve as a useful reference to determine the molecular identity of progenitors used in future preclinical and clinical studies.
Collapse
Affiliation(s)
- Devaveena Dey
- Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute, Institute of Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
Heart failure is a major cause of mortality worldwide with a steady increase in prevalence. There is currently no available cure beyond orthotopic heart transplantation, which for a number of reasons is an option only for a small fraction of all patients. Considerable hope has therefore been placed on the possibility of treating a failing heart by replacing lost cardiomyocytes, either through transplantation of various types of stem cells or by boosting endogenous regenerative mechanisms in the heart. Here, we review the current status of stem and progenitor cell-based therapies for heart disease. We discuss the pros and cons of different stem and progenitor cell types that can be considered for transplantation and describe recent advances in the understanding of how cardiomyocytes normally differentiate and how these cells can be generated from more immature cells ex vivo. Finally, we consider the possibility of activation of endogenous stem and progenitor cells to treat heart failure.
Collapse
Affiliation(s)
- E M Hansson
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| | | |
Collapse
|
238
|
Li J, Ezzelarab MB, Cooper DKC. Do mesenchymal stem cells function across species barriers? Relevance for xenotransplantation. Xenotransplantation 2013; 19:273-85. [PMID: 22978461 DOI: 10.1111/xen.12000] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Allogeneic mesenchymal stem (stromal) cells (MSC) are a promising therapy for various pathological conditions. Genetically modified pig MSC have been demonstrated to downregulate the human T-cell response to pig antigens in vitro. Before genetically modified pig MSC can be used clinically, however, evidence needs to be provided to indicate whether they will survive in a human (xenogeneic) host. LITERATURE SEARCH AND RESULTS A literature search through the end of 2011 identified 94 reports of the in vivo cross-species administration of MSC in a variety of experimental models. The majority (n = 89) involved the use of human MSC in various other species, with an occasional study using pig, rat, or guinea-pig MSC. When human MSC were used, they were largely derived from the bone marrow, adipose tissue, or umbilical cord blood. The routes of administration were varied, although almost half of the studies utilized the intravenous route. In 88 experiments (93.6%), there was evidence that the MSC engrafted and functioned across the species barrier, and in only six cases (6.4%) was there evidence of failure to function. Importantly, MSC function was confirmed in several different cross-species models. For example, human MSC functioned in no fewer than seven different recipient species. CONCLUSIONS The data provided by this literature search strengthen the hypothesis that pig MSC will function satisfactorily in a different species, for example, humans. The data also suggest that our own in vitro observations on the efficacy of pig MSC in downregulating the strength of the human T-cell response to pig antigens will likely be reproduced in vivo in pre-clinical large animal models and in clinical trials.
Collapse
Affiliation(s)
- Jiang Li
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
239
|
Ahmed LA. Stem cells and cardiac repair: alternative and multifactorial approaches. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
240
|
Zotarelli Filho IJ, Frascino LF, Greco OT, Araújo JDD, Bilaqui A, Kassis EN, Ardito RV, Bonilla-Rodriguez GO. A clinical and radiographic evaluation of the management of periodontal osseous defects with alloplast and platelet rich plasma. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
241
|
Kidney. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
242
|
Sakamoto M, Nakamura T, Torimura T, Iwamoto H, Masuda H, Koga H, Abe M, Hashimoto O, Ueno T, Sata M. Transplantation of endothelial progenitor cells ameliorates vascular dysfunction and portal hypertension in carbon tetrachloride-induced rat liver cirrhotic model. J Gastroenterol Hepatol 2013; 28:168-78. [PMID: 22849788 DOI: 10.1111/j.1440-1746.2012.07238.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2012] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIM In cirrhosis, sinusoidal endothelial cell injury results in increased endothelin-1 (ET-1) and decreased nitric oxide synthase (NOS) activity, leading to portal hypertension. However, the effects of transplanted endothelial progenitor cells (EPCs) on the cirrhotic liver have not yet been clarified. We investigated whether EPC transplantation reduces portal hypertension. METHODS Cirrhotic rats were created by the administration of carbon tetrachloride (CCl(4) ) twice weekly for 10 weeks. From week 7, rat bone marrow-derived EPCs were injected via the tail vein in this model once a week for 4 weeks. Endothelial NOS (eNOS), vascular endothelial growth factor (VEGF) and caveolin expressions were examined by Western blots. Hepatic tissue ET-1 was measured by a radioimmunoassay (RIA). Portal venous pressure, mean aortic pressure, and hepatic blood flow were measured. RESULTS Endothelial progenitor cell transplantation reduced liver fibrosis, α-smooth muscle actin-positive cells, caveolin expression, ET-1 concentration and portal venous pressure. EPC transplantation increased hepatic blood flow, protein levels of eNOS and VEGF. Immunohistochemical analyses of eNOS and isolectin B4 demonstrated that the livers of EPC-transplanted animals had markedly increased vascular density, suggesting reconstitution of sinusoidal blood vessels with endothelium. CONCLUSIONS Transplantation of EPCs ameliorates vascular dysfunction and portal hypertension, suggesting this treatment may provide a new approach in the therapy of portal hypertension with liver cirrhosis.
Collapse
Affiliation(s)
- Masaharu Sakamoto
- Department of Gastroenterology and Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
2D and 3D self-assembling nanofiber hydrogels for cardiomyocyte culture. BIOMED RESEARCH INTERNATIONAL 2012; 2013:285678. [PMID: 23573513 PMCID: PMC3593039 DOI: 10.1155/2013/285678] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/09/2012] [Accepted: 10/12/2012] [Indexed: 01/22/2023]
Abstract
Collagen is a widely used biomaterial in cardiac tissue engineering studies. However, as a natural material, it suffers from variability between batches that can complicate the standardization of culture conditions. In contrast, synthetic materials are modifiable, have well-defined structures and more homogeneous batches can be produced. In this study, several collagen-like synthetic self-assembling nanofiber hydrogels were examined for their suitability for cardiomyocyte culture in 2D and 3D. Six different nanofiber coatings were used in the 2D format with neonatal rat cardiomyocytes (NRCs) and human embryonic stem-cell-derived cardiomyocytes (hESC-CMs). The viability, growth, and functionality of the 2D-cultured cardiomyocytes were evaluated. The best-performing nanofiber coatings were selected for 3D experiments. Hydrophilic pH-sensitive nanofiber hydrogel coassembled with hyaluronic acid performed best with both NRCs and hESC-CMs. Hydrophilic non-pH-sensitive nanofiber hydrogels supported the growth of NRCs; however, their ability to promote attachment and growth of hESC-CMs was limited. NRCs also grew on hydrophobic nanofiber hydrogels; however, the cell-supporting capacity of these hydrogels was inferior to that of the hydrophilic hydrogel materials. This is the first study demonstrating that hydrophilic self-assembling nanofiber hydrogels support the culture of both NRCs and hESC-CMs, which suggests that these biomaterials hold promise for cardiac tissue engineering.
Collapse
|
244
|
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells which have self-renewal capacity and differentiation potential into several mesenchymal lineages including bones, cartilages, adipose tissues and tendons. MSCs may repair tissue injuries and prevent immune cell activation and proliferation. Immunomodulation and secretion of growth factors by MSCs have led to realizing the true potential of MSC-based cell therapy. The use of MSCs as immunomodulators has been explored in cell/organ transplant, tissue repair, autoimmune diseases, and prevention of graft vs host disease (GVHD). This review focuses on the clinical applications of MSC-based cell therapy, with particular emphasis on islet transplantation for treating type I diabetes.
Collapse
Affiliation(s)
- Vaibhav Mundra
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38103, United States
| | | | | |
Collapse
|
245
|
Giordano C, Kuraitis D, Beanlands RSB, Suuronen EJ, Ruel M. Cell-based vasculogenic studies in preclinical models of chronic myocardial ischaemia and hibernation. Expert Opin Biol Ther 2012; 13:411-28. [PMID: 23256710 DOI: 10.1517/14712598.2013.748739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Coronary artery disease commonly leads to myocardial ischaemia and hibernation. Relevant preclinical models of these conditions are essential to evaluate new therapeutic options such as cell-based vasculogenic therapies. AREAS COVERED In this article, the authors first review basic concepts of myocardial ischaemia/hibernation and relevant techniques to assess myocardial viability. Then, preclinical models of chronic myocardial ischaemia and hibernation, induced by devices such as ameroid constrictors, Delrin stenosis, hydraulic occluders, and coils/stents are described. Lastly, the authors discuss cell-based vasculogenic therapy, and summarise studies conducted in large animal models of chronic myocardial ischaemia and hibernation. EXPERT OPINION Approximately one-third of patients with viable myocardium do not undergo revascularisation; however, this population is at high risk for cardiac events and would surely benefit from effective cell-based therapy. Because of the modest benefits in clinical studies, preclinical models accurately representing clinical myocardial ischemia/hibernation are necessary to better understand and appropriately direct regenerative therapy research.
Collapse
Affiliation(s)
- Céline Giordano
- University of Ottawa Heart Institute, Division of Cardiac Surgery, 40 Ruskin Street, Suite 3403, Ottawa, Ontario, K1Y 4W7, Canada
| | | | | | | | | |
Collapse
|
246
|
Guenoun J, Ruggiero A, Doeswijk G, Janssens RC, Koning GA, Kotek G, Krestin GP, Bernsen MR. In vivoquantitative assessment of cell viability of gadolinium or iron-labeled cells using MRI and bioluminescence imaging. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 8:165-74. [DOI: 10.1002/cmmi.1513] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 08/21/2012] [Accepted: 09/10/2012] [Indexed: 01/09/2023]
Affiliation(s)
- Jamal Guenoun
- Department of Radiology; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | - Alessandro Ruggiero
- Department of Radiology; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | - Gabriela Doeswijk
- Department of Radiology; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | - Roel C. Janssens
- Department of Genetics; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | - Gerben A. Koning
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | - Gyula Kotek
- Department of Radiology; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | - Gabriel P. Krestin
- Department of Radiology; Erasmus MC - University Medical Center Rotterdam; Rotterdam; The Netherlands
| | | |
Collapse
|
247
|
Chang CL, Leu S, Sung HC, Zhen YY, Cho CL, Chen A, Tsai TH, Chung SY, Chai HT, Sun CK, Yen CH, Yip HK. Impact of apoptotic adipose-derived mesenchymal stem cells on attenuating organ damage and reducing mortality in rat sepsis syndrome induced by cecal puncture and ligation. J Transl Med 2012; 10:244. [PMID: 23217183 PMCID: PMC3543276 DOI: 10.1186/1479-5876-10-244] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 11/28/2012] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND We tested whether apoptotic adipose-derived mesenchymal stem cells (A-ADMSCs) were superior to healthy (H)-ADMSCs at attenuating organ damage and mortality in sepsis syndrome following cecal ligation and puncture (CLP). METHODS Adult male rats were categorized into group 1 (sham control), group 2 (CLP), group 3 [CLP + H-ADMSC administered 0.5, 6, and 18 h after CLP], group 4 [CLP + A-ADMSC administered as per group 3]. RESULTS Circulating peak TNF-α level, at 6 h, was highest in groups 2 and 3, and higher in group 4 than group 1 (p < 0.0001). Immune reactivity (indicated by circulating and splenic helper-, cytoxic-, and regulatory-T cells) at 24 and 72 h exhibited the same pattern as TNF-α amongst the groups (all p < 0.0001). The mononuclear-cell early and late apoptosis level and organ damage parameters of liver (AST, ALT), kidney (creatinine) and lung (arterial oxygen saturation) also displayed a similar pattern to TNF-α levels (all p < 0.001). Protein levels of inflammatory (TNF-α, MMP-9, NF-κB, ICAM-1), oxidative (oxidized protein) and apoptotic (Bax, caspase-3, PARP) biomarkers were higher in groups 2 and 3 than group 1, whereas anti-apoptotic (Bcl-2) biomarker was lower in groups 2 and 3 than in group 1 but anti-oxidant (GR, GPx, HO-1, NQO-1) showed an opposite way of Bcl-2; these patterns were reversed for group 4 (all p < 0.001). Mortality was highest in group 3 and higher in group 2 than group 4 than group 1 (all p < 0.001). CONCLUSIONS A-ADMSC therapy protected major organs from damage and improved prognosis in rats with sepsis syndrome.
Collapse
Affiliation(s)
- Chia-Lo Chang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Welt FGP, Gallegos R, Connell J, Kajstura J, D'Amario D, Kwong RY, Coelho-Filho O, Shah R, Mitchell R, Leri A, Foley L, Anversa P, Pfeffer MA. Effect of cardiac stem cells on left-ventricular remodeling in a canine model of chronic myocardial infarction. Circ Heart Fail 2012; 6:99-106. [PMID: 23212553 DOI: 10.1161/circheartfailure.112.972273] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Regenerative medicine, including cell therapy, is a promising strategy for recovery of the damaged myocardium. C-kit-positive cardiac stem cells (CSCs) have been shown to improve myocardial function after ischemic injury in animal models and in early clinical experience. We used a chronic large animal model of myocardial infarction with substantial reductions in left-ventricular (LV) ejection fraction and adverse remodeling to examine the effect of late autologous CSC intramyocardial injection on long-term cardiac structure and function. METHODS AND RESULTS Thoracotomy and ligation of the proximal left anterior descending artery, additional diagonal branches, and atrial biopsy for CSC culture were performed in canines. Baseline cardiac MRI was performed at 6 weeks postinfarct followed by repeat thoracotomy for randomization to intramyocardial injection of CSCs (n=13) or vehicle alone (n=6). At 30 weeks postmyocardial infarction, repeat MRI was performed. Data were analyzed using nonparametric tests (Wilcoxon signed-rank and rank-sum tests). In control animals, LV end-systolic volume and end-diastolic volume increased from 6 to 30 weeks (median and interquartile range, 51.3 mL [43.3-57.4] to 76.1 mL [72.0-82.4]; P=0.03 and 78.5 mL [69.7-86.1] to 99.2 mL [97.1-100.4]; P=0.03). Left-ventricular ejection fraction declined further (35.2% [27.9-38.7] to 26.4% [22.0-31.0]; P=0.12). In the cell-treated animals, this late adverse LV remodeling was attenuated (LV end-systolic volume, 42.6 mL [38.5-50.5] to 56.1 mL [50.3-63.0]; P=0.01 versus control). There was a nonsignificant attenuation in the increase in LV end-diastolic volume (64.8 mL [60.7-71.3] to 83.5 mL [74.7-90.8]; P=0.14 versus control) and LV ejection fraction change over time differed (30.5% [28.4-33.4] to 32.9% [28.6-36.9]; P=0.04 versus control). CONCLUSIONS Intramyocardial injection of autologous CSCs in a late phase model of chronic infarction resulted in less increase in LV end-systolic volume and preservation of LV ejection fraction.
Collapse
|
249
|
Matheeussen V, Jungraithmayr W, De Meester I. Dipeptidyl peptidase 4 as a therapeutic target in ischemia/reperfusion injury. Pharmacol Ther 2012; 136:267-82. [DOI: 10.1016/j.pharmthera.2012.07.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 07/16/2012] [Indexed: 01/21/2023]
|
250
|
Jiang J, Yao P, Gu Y, Xu L, Xu J, Tan H. Adult rat mesenchymal stem cells delay denervated muscle atrophy. Cell Mol Neurobiol 2012; 32:1287-98. [PMID: 22777537 PMCID: PMC11498426 DOI: 10.1007/s10571-012-9853-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 04/24/2012] [Indexed: 12/13/2022]
Abstract
To evaluate the function of rat mesenchymal stem cells (rMSCs) on denervated gastrocnemius muscles and to address the role of ciliary neurotrophic factor (CNTF) in rMSCs, denervated Wistar rats were separately injected with culture media (sham control), CNTF protein, 2.5 × 10(5) siCNTF-treated rMSCs, 2.5 × 10(5) GFP-transfected rMSCs, or 2.5 × 10(5) untreated rMSCs. Muscle function was assessed at different time points post-surgery. Tibial nerve and gastrocnemius muscle samples were taken at 4, 8, and 12 weeks for histochemistry, and neuromuscular junction repair was also examined by electron microscopy. Fluorescence immunocytochemistry on tissue sections confirmed neurotrophin expression in rMSCs but with little evidence of neuronal differentiation. The engraftment of rMSCs significantly preserved the function of denervated gastrocnemius muscle based both on evaluation of muscle function and direct examination of muscle tissue. Further, the density and depth of the junctional folds were visibly reduced 12 weeks after surgery and transplantation, especially in control group. Knockdown of CNTF expression in rMSCs failed to block muscle preservation, although administration of CNTF protein alone inhibited muscle atrophy, which indicating that delivery of rMSCs could preserve gastrocnemius muscle function following denervation and post-junctional mechanisms involved in the repairing capability of rMSCs.
Collapse
Affiliation(s)
- Junjian Jiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040 China
| | - Ping Yao
- Department of Hand Surgery, Hang Zhou Plastic Surgery Hospital, Hangzhou, 310014 China
| | - Yudong Gu
- Department of Hand Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040 China
| | - Lei Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040 China
| | - Jianguang Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040 China
| | - Haitao Tan
- Orthopedics Department, The Eight Affiliated Hospital of Guangxi Medical University, No. 1 Zhongshan Middle Road, Guigang, 537100 China
| |
Collapse
|