101
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a broad overview of current trends in stem cell research and its applications in cardiovascular medicine. Researches on different stem cell sources, their inherent characteristics, and the limitations they have in medical applications are discussed. Additionally, uses of stem cells for both modeling and treating cardiovascular disease are discussed, taking note of the obstacles these engineered interventions must overcome to be clinically viable. RECENT FINDINGS Tissue engineering aims to replace dysfunctional tissues with engineered constructs. Stem cell technologies have been a great enabling factor in working toward this goal. Many tissue-engineered products are in development that utilize stem cell technology. Although promising, some refinement must be made to these constructs with respect to safety and functionality. A deeper understanding of basic differentiation and tissue developmental mechanisms is required to allow these engineered tissues to be translated into the clinic.
Collapse
Affiliation(s)
- Christopher W Anderson
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Molecular Cell Genetics and Developmental Biology Program, Yale University, New Haven, CT, 06510, USA
| | - Nicole Boardman
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Jiesi Luo
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Jinkyu Park
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Yibing Qyang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA.
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA.
- Yale Stem Cell Center, Yale University, New Haven, CT, 06510, USA.
- Department of Pathology, Yale University, New Haven, CT, 06510, USA.
| |
Collapse
|
102
|
The Use of Endothelial Progenitor Cells for the Regeneration of Musculoskeletal and Neural Tissues. Stem Cells Int 2017; 2017:1960804. [PMID: 28458693 PMCID: PMC5387841 DOI: 10.1155/2017/1960804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 03/12/2017] [Indexed: 12/18/2022] Open
Abstract
Endothelial progenitor cells (EPCs) derived from bone marrow and blood can differentiate into endothelial cells and promote neovascularization. In addition, EPCs are a promising cell source for the repair of various types of vascularized tissues and have been used in animal experiments and clinical trials for tissue repair. In this review, we focused on the kinetics of endogenous EPCs during tissue repair and the application of EPCs or stem cell populations containing EPCs for tissue regeneration in musculoskeletal and neural tissues including the bone, skeletal muscle, ligaments, spinal cord, and peripheral nerves. EPCs can be mobilized from bone marrow and recruited to injured tissue to contribute to neovascularization and tissue repair. In addition, EPCs or stem cell populations containing EPCs promote neovascularization and tissue repair through their differentiation to endothelial cells or tissue-specific cells, the upregulation of growth factors, and the induction and activation of endogenous stem cells. Human peripheral blood CD34(+) cells containing EPCs have been used in clinical trials of bone repair. Thus, EPCs are a promising cell source for the treatment of musculoskeletal and neural tissue injury.
Collapse
|
103
|
Tobin SW, Li SH, Li J, Wu J, Yeganeh A, Yu P, Weisel RD, Li RK. Dual roles for bone marrow-derived Sca-1 cells in cardiac function. FASEB J 2017; 31:2905-2915. [PMID: 28336524 DOI: 10.1096/fj.201601363rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/06/2017] [Indexed: 01/04/2023]
Abstract
Recruitment of stem cells from the bone marrow (BM) is an important aspect of cardiac healing that becomes inefficient with age. We investigated the role of young stem cell antigen 1 (Sca-1)-positive BM cells on the aged heart by microarray analysis after BM reconstitution. Sca-1+ and Sca-1- BM cells from young green fluorescent protein (GFP)-positive mice were used to reconstitute the BM of aged mice. Myocardial infarction (MI) was induced 3 mo later. GFP+ cells were more abundant in the BM, blood, and heart of Sca-1+ mice, which corresponded to preserved cardiac function after MI. At baseline, Sca-1+ BM reconstitution increased cardiac expression of serum response factor, vascular endothelial growth factor A, and myogenic genes, but reduced the expression of Il-1β. After MI, inflammation was identified as a key difference between Sca-1- and Sca-1+ groups, as cytokine expression and cell surface markers associated with inflammatory cells were up-regulated with Sca-1+ reconstitution. Mac-3 and F4/80 staining showed that the postinfarction heart was composed of a mixture of GFP+ (donor) macrophages, GFP- (host) macrophages, and GFP+ cells that did not contribute to the macrophage population. This study demonstrates that Sca-1+ BM cells regulate cardiac healing though an acute inflammatory response and also before injury by stimulating formation of a beneficial cardiac niche.-Tobin, S. W., Li, S.-H., Li, J., Wu, J., Yeganeh, A., Yu, P., Weisel, R. D., Li, R.-K. Dual roles for bone marrow-derived Sca-1 cells in cardiac function.
Collapse
Affiliation(s)
- Stephanie W Tobin
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shu-Hong Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jiao Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun Wu
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Azadeh Yeganeh
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Pan Yu
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Richard D Weisel
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; .,Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
104
|
Valarmathi MT, Fuseler JW, Davis JM, Price RL. A Novel Human Tissue-Engineered 3-D Functional Vascularized Cardiac Muscle Construct. Front Cell Dev Biol 2017; 5:2. [PMID: 28194397 PMCID: PMC5276820 DOI: 10.3389/fcell.2017.00002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/11/2017] [Indexed: 12/28/2022] Open
Abstract
Organ tissue engineering, including cardiovascular tissues, has been an area of intense investigation. The major challenge to these approaches has been the inability to vascularize and perfuse the in vitro engineered tissue constructs. Attempts to provide oxygen and nutrients to the cells contained in the biomaterial constructs have had varying degrees of success. The aim of this current study is to develop a three-dimensional (3-D) model of vascularized cardiac tissue to examine the concurrent temporal and spatial regulation of cardiomyogenesis in the context of postnatal de novo vasculogenesis during stem cell cardiac regeneration. In order to achieve the above aim, we have developed an in vitro 3-D functional vascularized cardiac muscle construct using human induced pluripotent stem cell-derived embryonic cardiac myocytes (hiPSC-ECMs) and human mesenchymal stem cells (hMSCs). First, to generate the prevascularized scaffold, human cardiac microvascular endothelial cells (hCMVECs) and hMSCs were co-cultured onto a 3-D collagen cell carrier (CCC) for 7 days under vasculogenic culture conditions. In this milieu, hCMVECs/hMSCs underwent maturation, differentiation, and morphogenesis characteristic of microvessels, and formed extensive plexuses of vascular networks. Next, the hiPSC-ECMs and hMSCs were co-cultured onto this generated prevascularized CCCs for further 7 or 14 days in myogenic culture conditions. Finally, the vascular and cardiac phenotypic inductions were analyzed at the morphological, immunological, biochemical, molecular, and functional levels. Expression and functional analyses of the differentiated cells revealed neo-angiogenesis and neo-cardiomyogenesis. Thus, our unique 3-D co-culture system provided us the apt in vitro functional vascularized 3-D cardiac patch that can be utilized for cellular cardiomyoplasty.
Collapse
Affiliation(s)
- Mani T Valarmathi
- Laboratory of Stem Cell Biology and Tissue Engineering, Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign Urbana, IL, USA
| | - John W Fuseler
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Jeffrey M Davis
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA
| | - Robert L Price
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina Columbia, SC, USA
| |
Collapse
|
105
|
Progenitor Cells for Arterial Repair: Incremental Advancements towards Therapeutic Reality. Stem Cells Int 2017; 2017:8270498. [PMID: 28232850 PMCID: PMC5292398 DOI: 10.1155/2017/8270498] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/18/2016] [Indexed: 02/08/2023] Open
Abstract
Coronary revascularization remains the standard treatment for obstructive coronary artery disease and can be accomplished by either percutaneous coronary intervention (PCI) or coronary artery bypass graft surgery. Considerable advances have rendered PCI the most common form of revascularization and improved clinical outcomes. However, numerous challenges to modern PCI remain, namely, in-stent restenosis and stent thrombosis, underscoring the importance of understanding the vessel wall response to injury to identify targets for intervention. Among recent promising discoveries, endothelial progenitor cells (EPCs) have garnered considerable interest given an increasing appreciation of their role in vascular homeostasis and their ability to promote vascular repair after stent placement. Circulating EPC numbers have been inversely correlated with cardiovascular risk, while administration of EPCs in humans has demonstrated improved clinical outcomes. Despite these encouraging results, however, advancing EPCs as a therapeutic modality has been hampered by a fundamental roadblock: what constitutes an EPC? We review current definitions and sources of EPCs as well as the proposed mechanisms of EPC-mediated vascular repair. Additionally, we discuss the current state of EPCs as therapeutic agents, focusing on endogenous augmentation and transplantation.
Collapse
|
106
|
Therapeutic Potential of Hematopoietic Stem Cell-Derived Exosomes in Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 998:221-235. [DOI: 10.1007/978-981-10-4397-0_15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
107
|
Chen S, Mao H, Tu P, Cheng N, Yu J. Fabrication of decellularized adipose tissue/alginate composite microspheres with pASCs encapsulation for tissue engineering. AIMS BIOENGINEERING 2017. [DOI: 10.3934/bioeng.2017.3.351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
108
|
Wu F, Wang C, Xu Y, Ma J. Model of electrical activity in cardiac tissue under electromagnetic induction. Sci Rep 2016; 6:28. [PMID: 28442705 PMCID: PMC5431370 DOI: 10.1038/s41598-016-0031-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/31/2016] [Indexed: 11/09/2022] Open
Abstract
Complex electrical activities in cardiac tissue can set up time-varying electromagnetic field. Magnetic flux is introduced into the Fitzhugh-Nagumo model to describe the effect of electromagnetic induction, and then memristor is used to realize the feedback of magnetic flux on the membrane potential in cardiac tissue. It is found that a spiral wave can be triggered and developed by setting specific initials in the media, that is to say, the media still support the survival of standing spiral waves under electromagnetic induction. Furthermore, electromagnetic radiation is considered on this model as external stimuli, it is found that spiral waves encounter breakup and turbulent electrical activities are observed, and it can give guidance to understand the occurrence of sudden heart disorder subjected to heavily electromagnetic radiation.
Collapse
Affiliation(s)
- Fuqiang Wu
- Department of Physics, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Chunni Wang
- Department of Physics, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Ying Xu
- Department of Physics, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Jun Ma
- Department of Physics, Lanzhou University of Technology, Lanzhou, 730050, China.
| |
Collapse
|
109
|
Kornicka K, Marycz K, Marędziak M, Tomaszewski KA, Nicpoń J. The effects of the DNA methyltranfserases inhibitor 5-Azacitidine on ageing, oxidative stress and DNA methylation of adipose derived stem cells. J Cell Mol Med 2016; 21:387-401. [PMID: 27998022 PMCID: PMC5264131 DOI: 10.1111/jcmm.12972] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 08/10/2016] [Indexed: 12/23/2022] Open
Abstract
Human adipose tissue is a great source of adult mesenchymal stem cells (MSCs) which are recognized from their ability to self‐renew and differentiation into multiple lineages. MSCs have promised a vast therapeutic potential in treatment many diseases including tissue injury and immune disorders. However, their regenerative potential profoundly depends on patients’ age. Age‐related deterioration of MSC is associated with cellular senescence mainly caused by increased DNA methylation status, accumulation of oxidative stress factors and mitochondria dysfunction. We found that DNA methyltransferase (DNMT) inhibitor i.e. 5‐Azacytidine (5‐AZA) reversed the aged phenotype of MSCs. Proliferation rate of cells cultured with 5‐AZA was increased while the accumulation of oxidative stress factors and DNA methylation status were decreased. Simultaneously the mRNA levels of TET proteins involved in demethylation process were elevated in those cells. Moreover, cells treated with 5‐AZA displayed reduced reactive oxygen species (ROS) accumulation, ameliorated superoxide dismutase activity and increased BCL‐2/BAX ratio in comparison to control group. Our results indicates that, treating MSCs with 5‐AZA can be justified therapeutic intervention, that can slow‐down and even reverse aged‐ related degenerative changes in those cells.
Collapse
Affiliation(s)
- Katarzyna Kornicka
- Faculty of Biology, University of Environmental and Life Sciences, Wrocław, Poland.,Wroclaw Research Centre EIT+, Wrocław, Poland
| | - Krzysztof Marycz
- Faculty of Biology, University of Environmental and Life Sciences, Wrocław, Poland.,Wroclaw Research Centre EIT+, Wrocław, Poland
| | - Monika Marędziak
- Faculty of Veterinary Medicine, University of Environmental and Life Sciences, Wrocław, Poland
| | | | - Jakub Nicpoń
- Department of Surgery, Faculty of Veterinary Medicine, University of Environmental and Life Sciences Wroclaw, Wroclaw, Poland
| |
Collapse
|
110
|
Kaigler D, Krebsbach PH, Wang Z, West ER, Horger K, Mooney DJ. Transplanted Endothelial Cells Enhance Orthotopic Bone Regeneration. J Dent Res 2016; 85:633-7. [PMID: 16798864 DOI: 10.1177/154405910608500710] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The aim of this study was to determine if endothelial cells could enhance bone marrow stromal-cell-mediated bone regeneration in an osseous defect. Using poly-lactide-co-glycolide scaffolds as cell carriers, we transplanted bone marrow stromal cells alone or with endothelial cells into 8.5-mm calvarial defects created in nude rats. Histological analyses of blood vessel and bone formation were performed, and microcomputed tomography (μCT) was used to assess mineralized bone matrix. Though the magnitude of the angiogenic response between groups was the same, μCT analysis revealed earlier mineralization of bone in the co-transplantation condition. Ultimately, there was a significant increase (40%) in bone formation in the co-transplantation group (33 ± 2%), compared with the transplantation of bone marrow stromal cells alone (23 ± 3%). Analysis of these data demonstrates that, in an orthotopic site, transplanted endothelial cells can influence the bone-regenerative capacity of bone marrow stromal cells.
Collapse
Affiliation(s)
- D Kaigler
- Dept. of Periodontics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
111
|
Chou KJ, Lee PT, Chen CL, Hsu CY, Huang WC, Huang CW, Fang HC. CD44 fucosylation on mesenchymal stem cell enhances homing and macrophage polarization in ischemic kidney injury. Exp Cell Res 2016; 350:91-102. [PMID: 27871849 DOI: 10.1016/j.yexcr.2016.11.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 01/14/2023]
Abstract
The lack of homing ability possibly reduces the healing potential of bone-marrow-derived mesenchymal stem cells (MSCs). Therefore, transforming native CD44 on MSCs into a hematopoietic cell E-/L-selectin ligand (HCELL) that possesses potent E-selectin affinity might enhance the homing and regenerative abilities of MSCs. Through fucosyltransferase VI (FTVI) transfection, MSCs were fucosylated on N-glycans of CD44 to become HCELL positive, thus interacting with E-selectin on injured endothelial cells. HCELL expression facilitated MSC homing in kidneys within 24h after injury and reduced lung stasis. An in vitro adhesion assay revealed that transfection enhanced the association between MSCs and hypoxic endothelial cells. In mice treated with HCELL-positive MSCs, the injured kidneys exhibited clusters of homing MSCs, whereas MSCs were rarely observed in mouse kidneys treated with HCELL-negative MSCs. Most MSCs were initially localized at the renal capsule, and some MSCs later migrated inward between tubules. Most homing MSCs were in close contact with inflammatory cells without tubular transdifferentiation. Furthermore, HCELL-positive MSCs substantially alleviated renal injury, partly by enhancing the polarization of infiltrating macrophages. In conclusion, engineering the glycan of CD44 on MSCs through FTVI transfection might enhance renotropism and the regenerating ability of MSCs in ischemic kidney injury.
Collapse
Affiliation(s)
- Kang-Ju Chou
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Tsang Lee
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Liang Chen
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Yang Hsu
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Chieh Huang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chien-Wei Huang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Hua-Chang Fang
- Division of Nephrology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
112
|
O'Gallagher K, Astroulakis Z, Sirker A, Hill JM. Concepts of Cell Therapy and Myocardial Regeneration. Interv Cardiol 2016. [DOI: 10.1002/9781118983652.ch29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Kevin O'Gallagher
- Department of Cardiology; King's College Hospital NHS Foundation Trust; London UK
| | | | - Alex Sirker
- Department of Cardiology; UCLH and St Bartholomew's Hospital; London UK
| | - Jonathan M. Hill
- Department of Cardiology; King's College Hospital NHS Foundation Trust; London UK
| |
Collapse
|
113
|
Yellamilli A, van Berlo JH. The Role of Cardiac Side Population Cells in Cardiac Regeneration. Front Cell Dev Biol 2016; 4:102. [PMID: 27679798 PMCID: PMC5020051 DOI: 10.3389/fcell.2016.00102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022] Open
Abstract
The heart has a limited ability to regenerate. It is important to identify therapeutic strategies that enhance cardiac regeneration in order to replace cardiomyocytes lost during the progression of heart failure. Cardiac progenitor cells are interesting targets for new regenerative therapies because they are self-renewing, multipotent cells located in the heart. Cardiac side population cells (cSPCs), the first cardiac progenitor cells identified in the adult heart, have the ability to differentiate into cardiomyocytes, endothelial cells, smooth muscle cells, and fibroblasts. They become activated in response to cardiac injury and transplantation of cSPCs into the injured heart improves cardiac function. In this review, we will discuss the current literature on the progenitor cell properties and therapeutic potential of cSPCs. This body of work demonstrates the great promise cSPCs hold as targets for new regenerative strategies.
Collapse
Affiliation(s)
- Amritha Yellamilli
- Stem Cell Institute, University of MinnesotaMinneapolis, MN, USA; Lillehei Heart Institute, University of MinnesotaMinneapolis, MN, USA; Department of Integrative Biology and Physiology, University of MinnesotaMinneapolis, MN, USA
| | - Jop H van Berlo
- Stem Cell Institute, University of MinnesotaMinneapolis, MN, USA; Lillehei Heart Institute, University of MinnesotaMinneapolis, MN, USA; Department of Integrative Biology and Physiology, University of MinnesotaMinneapolis, MN, USA; Department of Medicine/Cardiology, University of MinnesotaMinneapolis, MN, USA
| |
Collapse
|
114
|
Abstract
Mesenchymal stem cells (MSCs) have great potential as a source of cells for cell-based therapy because of their ability for self-renewal and differentiation into functional cells. Moreover, matrix metalloproteinases (MMPs) have a critical role in the differentiation of MSCs into different lineages. MSCs also interact with exogenous MMPs at their surface, and regulate the pericellular localization of MMP activities. The fate of MSCs is regulated by specific MMPs associated with a key cell lineage. Recent reports suggest the integration of MMPs in the differentiation, angiogenesis, proliferation, and migration of MSCs. These interactions are not fully understood and warrant further investigation, especially for their application as therapeutic tools to treat different diseases. Therefore, overexpression of a single MMP or tissue-specific inhibitor of metalloproteinase in MSCs may promote transdifferentiation into a specific cell lineage, which can be used for the treatment of some diseases. In this review, we critically discuss the identification of various MMPs and the signaling pathways that affect the differentiation, migration, angiogenesis, and proliferation of MSCs.
Collapse
Affiliation(s)
- Sami G Almalki
- Department of Clinical and Translational Science, Creighton University School of Medicine, CRISS II, Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, CRISS II, Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
115
|
Nyström A, Bornert O, Kühl T. Cell therapy for basement membrane-linked diseases. Matrix Biol 2016; 57-58:124-139. [PMID: 27609402 DOI: 10.1016/j.matbio.2016.07.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/02/2016] [Accepted: 07/07/2016] [Indexed: 12/14/2022]
Abstract
For most disorders caused by mutations in genes encoding basement membrane (BM) proteins, there are at present only limited treatment options available. Genetic BM-linked disorders can be viewed as especially suited for treatment with cell-based therapy approaches because the proteins that need to be restored are located in the extracellular space. In consequence, complete and permanent engraftment of cells does not necessarily have to occur to achieve substantial causal therapeutic effects. For these disorders cells can be used as transient vehicles for protein replacement. In addition, it is becoming evident that BM-linked genetic disorders are modified by secondary diseases mechanisms. Cell-based therapies have also the ability to target such disease modifying mechanisms. Thus, cell therapies can simultaneously provide causal treatment and symptomatic relief, and accordingly hold great potential for treatment of BM-linked disorders. However, this potential has for most applications and diseases so far not been realized. Here, we will present the state of cell therapies for BM-linked diseases. We will discuss use of both pluripotent and differentiated cells, the limitation of the approaches, their challenges, and the way forward to potential wider implementation of cell therapies in the clinics.
Collapse
Affiliation(s)
- Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany.
| | - Olivier Bornert
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Tobias Kühl
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
116
|
Chhokar V, Tucker AL. Angiogenesis: Basic Mechanisms and Clinical Applications. Semin Cardiothorac Vasc Anesth 2016. [DOI: 10.1177/108925320300700304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development and maintenance of an adequate vascular supply is critical for the viability of normal and neoplastic tissues. Angiogenesis, the development of new blood vessels from preexisting capillary networks, plays an important role in a number of physiologic and pathologic processes, including reproduction, wound repair, inflammatory diseases, and tumor growth. Angiogenesis involves sequential steps that are triggered in response to angiogenic growth factors released by inflammatory, mesenchymal, or tumor cells that act as ligands for endothelial cell receptor tyrosine kinases. Stimulated endothelial cells detach from neighboring cells and migrate, proliferate, and form tubes. The immature tubes are subsequently invested and stabilized by pericytes or smooth muscle cells. Angiogenesis depends upon complex interactions among various classes of molecules, including adhesion molecules, proteases, structural proteins, cell surface receptors, and growth factors. The therapeutic manipulation of angiogenesis targeted against ischemic and neoplastic diseases has been investigated in preclinical animal models and in clinical trials. Proangiogenic trials that have stimulated vessel growth in ischemic coronary or peripheral tissues through expression, delivery, or stimulated release of growth factors have shown efficacy in animal models and mixed results in human clinical trials. Antiangiogenic trials have used strategies to block the function of molecules critical for new vessel growth or maturation in the treatment of a variety of malignancies, mostly with results less encouraging than those seen in preclinical models. Pro-and antiangiogenic clinical trials demonstrate that strategies for optimal drug delivery, dosing schedules, patient selection, and endpoint measurements need further investigation and refinement before the therapeutic manipulation of angiogenesis will realize its full clinical potential.
Collapse
Affiliation(s)
- Vikram Chhokar
- Department of Internal Medicine, Salem VA Health System, Roanoke, Virginia
| | - Amy L. Tucker
- Department of Internal Medicine, Cardiovascular Division; Cardiovascular Research Center; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
117
|
Medhekar SK, Shende VS, Chincholkar AB. Recent Stem Cell Advances: Cord Blood and Induced Pluripotent Stem Cell for Cardiac Regeneration- a Review. Int J Stem Cells 2016; 9:21-30. [PMID: 27426082 PMCID: PMC4961100 DOI: 10.15283/ijsc.2016.9.1.21] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 03/07/2016] [Indexed: 12/14/2022] Open
Abstract
Stem cells are primitive self renewing undifferentiated cell that can be differentiated into various types of specialized cells like nerve cell, skin cells, muscle cells, intestinal tissue, and blood cells. Stem cells live in bone marrow where they divide to make new blood cells and produces peripheral stem cells in circulation. Under proper environment and in presence of signaling molecules stem cells begin to develop into specialized tissues and organs. These unique characteristics make them very promising entities for regeneration of damaged tissue. Day by day increase in incidence of heart diseases including left ventricular dysfunction, ischemic heart disease (IHD), congestive heart failure (CHF) are the major cause of morbidity and mortality. However infracted tissue cannot regenerate into healthy tissue. Heart transplantation is only the treatment for such patient. Due to limitation of availability of donor for organ transplantation, a focus is made for alternative and effective therapy to treat such condition. In this review we have discussed the new advances in stem cells such as use of cord stem cells and iPSC technology in cardiac repair. Future approach of CB cells was found to be used in tissue repair which is specifically observed for improvement of left ventricular function and myocardial infarction. Here we have also focused on how iPSC technology is used for regeneration of cardiomyocytes and intiating neovascularization in myocardial infarction and also for study of pathophysiology of various degenerative diseases and genetic disease in research field.
Collapse
Affiliation(s)
| | - Vikas Suresh Shende
- Department of Pharmacology, Satara College of Pharmacy, Degaon, Satara (MH), India
| | | |
Collapse
|
118
|
Marędziak M, Tomaszewski K, Polinceusz P, Lewandowski D, Marycz K. Static magnetic field enhances the viability and proliferation rate of adipose tissue-derived mesenchymal stem cells potentially through activation of the phosphoinositide 3-kinase/Akt (PI3K/Akt) pathway. Electromagn Biol Med 2016; 36:45-54. [PMID: 27367918 DOI: 10.3109/15368378.2016.1149860] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The aim of this work was to investigate the effects of 0.5T static magnetic field (sMF) on the viability and proliferation rate of human adipose-derived mesenchymal stromal stem cells (hASCs) via activation of the phosphoinositide 3-kinase/Akt (PI3K/Akt) signaling pathway. In a 7-d culture we examined cell growth kinetic and population doubling time (PDT). We also examined cell morphology and the cellular senescence markers level. Exposure to sMF enhanced the viability of these cells. However, the effect was blocked by treating the cells with LY294002, a P13K inhibitor. We compared this effect by Western Blot analysis of Akt protein expression. We also examined whether the cell response on sMF stimulation is dependent on integrin engagement and we measured integrin gene expression. Our results suggest that stimulation using sMF is a viable method to improve hASC viability. sMF is involved in mechanisms associated with controlling cell proliferative potential signaling events.
Collapse
Affiliation(s)
- Monika Marędziak
- a Faculty of Veterinary Medicine , University of Environmental and Life Sciences , Wrocław , Poland.,b Faculty of Biology , University of Environmental and Life Sciences , Wrocław , Poland
| | - Krzysztof Tomaszewski
- c Department of Anatomy , Jagiellonian University Medical College , Krakow , Poland.,d Department of Orthopaedics and Trauma Surgery , 5th Military Clinical Hospital and Polyclinic , Krakow , Poland
| | - Paulina Polinceusz
- e Faculty of Biotechnology , Laboratory of Cytobiochemistry, University of Wroclaw , Wrocław , Poland
| | - Daniel Lewandowski
- f Institute of Materials Science and Applied Mechanics, Wroclaw University of Technology , Wrocław , Poland
| | - Krzysztof Marycz
- b Faculty of Biology , University of Environmental and Life Sciences , Wrocław , Poland
| |
Collapse
|
119
|
Fang JS, Gritz EC, Marcelo KL, Hirschi KK. Isolation of Murine Embryonic Hemogenic Endothelial Cells. J Vis Exp 2016. [PMID: 27341393 PMCID: PMC4927799 DOI: 10.3791/54150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The specification of hemogenic endothelial cells from embryonic vascular endothelium occurs during brief developmental periods within distinct tissues, and is necessary for the emergence of definitive HSPC from the murine extra embryonic yolk sac, placenta, umbilical vessels, and the embryonic aorta-gonad-mesonephros (AGM) region. The transient nature and small size of this cell population renders its reproducible isolation for careful quantification and experimental applications technically difficult. We have established a fluorescence-activated cell sorting (FACS)-based protocol for simultaneous isolation of hemogenic endothelial cells and HSPC during their peak generation times in the yolk sac and AGM. We demonstrate methods for dissection of yolk sac and AGM tissues from mouse embryos, and we present optimized tissue digestion and antibody conjugation conditions for maximal cell survival prior to identification and retrieval via FACS. Representative FACS analysis plots are shown that identify the hemogenic endothelial cell and HSPC phenotypes, and describe a methylcellulose-based assay for evaluating their blood forming potential on a clonal level.
Collapse
Affiliation(s)
- Jennifer S Fang
- Departments of Medicine, Genetics and Biomedical Engineering, Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, Yale Stem Cell Center, Yale University School of Medicine
| | - Emily C Gritz
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Yale University School of Medicine
| | - Kathrina L Marcelo
- Department of Molecular and Cellular Biology, Baylor College of Medicine
| | - Karen K Hirschi
- Departments of Medicine, Genetics and Biomedical Engineering, Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, Yale Stem Cell Center, Yale University School of Medicine;
| |
Collapse
|
120
|
Allameh A, Jazayeri M, Adelipour M. In Vivo Vascularization of Endothelial Cells Derived from Bone Marrow Mesenchymal Stem Cells in SCID Mouse Model. CELL JOURNAL 2016; 18:179-88. [PMID: 27540522 PMCID: PMC4988416 DOI: 10.22074/cellj.2016.4312] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 10/01/2015] [Indexed: 12/29/2022]
Abstract
Objective In vivo and in vitro stem cell differentiation into endothelial cells is a promising
area of research for tissue engineering and cell therapy.
Materials and Methods We induced human mesenchymal stem cells (MSCs) to differentiate to endothelial cells that had the ability to form capillaries on an extracellular matrix
(ECM) gel. Thereafter, the differentiated endothelial cells at early stage were characterized
by expression of specific markers such as von Willebrand factor (vWF), vascular endothelial
growth factor (VEGF) receptor 2, and CD31. In this experimental model, the endothelial
cells were transplanted into the groins of severe combined immunodeficiency (SCID) mice.
After 30 days, we obtained tissue biopsies from the transplantation sites. Biopsies were
processed for histopathological and double immunohistochemistry (DIHC) staining.
Results Endothelial cells at the early stage of differentiation expressed endothelial markers. Hematoxylin and eosin (H&E) staining, in addition to DIHC demonstrated homing of
the endothelial cells that underwent vascularization in the injected site.
Conclusion The data clearly showed that endothelial cells at the early stage of differentiation underwent neovascularization in vivo in SCID mice. Endothelial cells at their early
stage of differentiation have been proven to be efficient for treatment of diseases with
impaired vasculogenesis.
Collapse
Affiliation(s)
- Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Jazayeri
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Biochemistry, School of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Adelipour
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
121
|
Oxygen cycling to improve survival of stem cells for myocardial repair: A review. Life Sci 2016; 153:124-31. [PMID: 27091653 DOI: 10.1016/j.lfs.2016.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/28/2016] [Accepted: 04/08/2016] [Indexed: 02/08/2023]
Abstract
Heart disease represents the leading cause of death among Americans. There is currently no clinical treatment to regenerate viable myocardium following myocardial infarction, and patients may suffer progressive deterioration and decreased myocardial function from the effects of remodeling of the necrotic myocardium. New therapeutic strategies hold promise for patients who suffer from ischemic heart disease by directly addressing the restoration of functional myocardium following death of cardiomyocytes. Therapeutic stem cell transplantation has shown modest benefit in clinical human trials with decreased fibrosis and increased functional myocardium. Moreover, autologous transplantation holds the potential to implement these therapies while avoiding the immunomodulation concerns of heart transplantation. Despite these benefits, stem cell therapy has been characterized by poor survival and low engraftment of injected stem cells. The hypoxic tissue environment of the ischemic/infracting myocardium impedes stem cell survival and engraftment in myocardial tissue. Hypoxic preconditioning has been suggested as a viable strategy to increase hypoxic tolerance of stem cells. A number of in vivo and in vitro studies have demonstrated improved stem cell viability by altering stem cell secretion of protein signals and up-regulation of numerous paracrine signaling pathways that affect inflammatory, survival, and angiogenic signaling pathways. This review will discuss both the mechanisms of hypoxic preconditioning as well as the effects of hypoxic preconditioning in different cell and animal models, examining the pitfalls in current research and the next steps into potentially implementing this methodology in clinical research trials.
Collapse
|
122
|
Shafiq M, Jung Y, Kim SH. Insight on stem cell preconditioning and instructive biomaterials to enhance cell adhesion, retention, and engraftment for tissue repair. Biomaterials 2016; 90:85-115. [PMID: 27016619 DOI: 10.1016/j.biomaterials.2016.03.020] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/09/2016] [Accepted: 03/13/2016] [Indexed: 12/13/2022]
Abstract
Stem cells are a promising solution for the treatment of a variety of diseases. However, the limited survival and engraftment of transplanted cells due to a hostile ischemic environment is a bottleneck for effective utilization and commercialization. Within this environment, the majority of transplanted cells undergo apoptosis prior to participating in lineage differentiation and cellular integration. Therefore, in order to maximize the clinical utility of stem/progenitor cells, strategies must be employed to increase their adhesion, retention, and engraftment in vivo. Here, we reviewed key strategies that are being adopted to enhance the survival, retention, and engraftment of transplanted stem cells through the manipulation of both the stem cells and the surrounding environment. We describe how preconditioning of cells or cell manipulations strategies can enhance stem cell survival and engraftment after transplantation. We also discuss how biomaterials can enhance the function of stem cells for effective tissue regeneration. Biomaterials can incorporate or mimic extracellular function (ECM) function and enhance survival or differentiation of transplanted cells in vivo. Biomaterials can also promote angiogenesis, enhance engraftment and differentiation, and accelerate electromechanical integration of transplanted stem cells. Insight gained from this review may direct the development of future investigations and clinical trials.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Korea University of Science and Technology, 176 Gajeong-dong, Yuseong-gu, Daejeon, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul 130-650, Republic of Korea
| | - Youngmee Jung
- Korea University of Science and Technology, 176 Gajeong-dong, Yuseong-gu, Daejeon, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul 130-650, Republic of Korea
| | - Soo Hyun Kim
- Korea University of Science and Technology, 176 Gajeong-dong, Yuseong-gu, Daejeon, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul 130-650, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 136-701, Republic of Korea.
| |
Collapse
|
123
|
Cellular Origins of Regenerating Nodules and Malignancy in the FAH Model of Liver Injury after Bone Marrow Cell Transplantation. Stem Cells Int 2016; 2016:5791317. [PMID: 26962307 PMCID: PMC4709791 DOI: 10.1155/2016/5791317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/04/2015] [Indexed: 12/02/2022] Open
Abstract
In previous reports, we and other groups have shown that proliferating hepatocytes are formed by the fusion of donor hematopoietic cells with host hepatocytes in the Fah−/− model. Thus, it would be interesting to determine whether cell fusion occurs during malignancy. However, it is difficult to demonstrate such processes using this model. Therefore, we established a new strain to study the processes of regenerating nodules and malignancy and their origins. The FAH−/− mouse model was crossed with the ROSAnZ strain and their offspring was genotyped for FAH−/− and ROSAnZ mutations to create a new strain (Fah−/−-ROSAnZ). Using this strain as recipients, we performed bone marrow transplantation experiments. As a result, we could not demonstrate the presence of any epithelial cells except hepatocytes that were of donor origin in regenerating tissue, and no evidence of cell fusion was found in tumors. The hepatic malignancy was of host origin in these mice. There was higher expression of extracellular matrix proteins and more inflammatory cells in liver tumor nodules than in regenerating normal liver nodules. Hepatocytes generated by fusion with bone marrow cells did not form malignant tumors. Extracellular matrix and inflammatory cells had significantly accumulated in liver tumors.
Collapse
|
124
|
Abdelwahid E, Kalvelyte A, Stulpinas A, de Carvalho KAT, Guarita-Souza LC, Foldes G. Stem cell death and survival in heart regeneration and repair. Apoptosis 2016; 21:252-68. [PMID: 26687129 PMCID: PMC5200890 DOI: 10.1007/s10495-015-1203-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are major causes of mortality and morbidity. Cardiomyocyte apoptosis disrupts cardiac function and leads to cardiac decompensation and terminal heart failure. Delineating the regulatory signaling pathways that orchestrate cell survival in the heart has significant therapeutic implications. Cardiac tissue has limited capacity to regenerate and repair. Stem cell therapy is a successful approach for repairing and regenerating ischemic cardiac tissue; however, transplanted cells display very high death percentage, a problem that affects success of tissue regeneration. Stem cells display multipotency or pluripotency and undergo self-renewal, however these events are negatively influenced by upregulation of cell death machinery that induces the significant decrease in survival and differentiation signals upon cardiovascular injury. While efforts to identify cell types and molecular pathways that promote cardiac tissue regeneration have been productive, studies that focus on blocking the extensive cell death after transplantation are limited. The control of cell death includes multiple networks rather than one crucial pathway, which underlies the challenge of identifying the interaction between various cellular and biochemical components. This review is aimed at exploiting the molecular mechanisms by which stem cells resist death signals to develop into mature and healthy cardiac cells. Specifically, we focus on a number of factors that control death and survival of stem cells upon transplantation and ultimately affect cardiac regeneration. We also discuss potential survival enhancing strategies and how they could be meaningful in the design of targeted therapies that improve cardiac function.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, 303 E. Chicago Ave., Tarry 14-725, Chicago, IL, 60611, USA.
| | - Audrone Kalvelyte
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Aurimas Stulpinas
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Katherine Athayde Teixeira de Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pequeno Príncipe Faculty, Pelé Pequeno Príncipe Institute, Curitiba, Paraná, 80250-200, Brazil
| | - Luiz Cesar Guarita-Souza
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Parana, Curitiba, Paraná, 80215-901, Brazil
| | - Gabor Foldes
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
125
|
Zou T, Fan J, Fartash A, Liu H, Fan Y. Cell-based strategies for vascular regeneration. J Biomed Mater Res A 2016; 104:1297-314. [PMID: 26864677 DOI: 10.1002/jbm.a.35660] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 01/17/2016] [Accepted: 01/19/2016] [Indexed: 01/12/2023]
Abstract
Vascular regeneration is known to play an essential role in the repair of injured tissues mainly through accelerating the repair of vascular injury caused by vascular diseases, as well as the recovery of ischemic tissues. However, the clinical vascular regeneration is still challenging. Cell-based therapy is thought to be a promising strategy for vascular regeneration, since various cells have been identified to exert important influences on the process of vascular regeneration such as the enhanced endothelium formation on the surface of vascular grafts, and the induction of vessel-like network formation in the ischemic tissues. Here are a vast number of diverse cell-based strategies that have been extensively studied in vascular regeneration. These strategies can be further classified into three main categories, including cell transplantation, construction of tissue-engineered grafts, and surface modification of scaffolds. Cells used in these strategies mainly refer to terminally differentiated vascular cells, pluripotent stem cells, multipotent stem cells, and unipotent stem cells. The aim of this review is to summarize the reported research advances on the application of various cells for vascular regeneration, yielding insights into future clinical treatment for injured tissue/organ.
Collapse
Affiliation(s)
- Tongqiang Zou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, People's Republic of China
| | - Jiabing Fan
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, 90095
| | - Armita Fartash
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, 90095
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, People's Republic of China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, People's Republic of China.,National Research Center for Rehabilitation Technical Aids, Beijing, 100176, People's Republic of China
| |
Collapse
|
126
|
Kort EJ, Croskey L, Scibienski T, Rajasekaran S, Jovinge S. Circulating Progenitor Cells and Childhood Cardiovascular Disease. Pediatr Cardiol 2016; 37:225-31. [PMID: 26554720 DOI: 10.1007/s00246-015-1300-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/20/2015] [Indexed: 12/18/2022]
Abstract
Circulating progenitor cells have been extensively studied in the context of heart disease in adults. In these patients, they have been demonstrated to be markers of myocardial injury and recovery as well as potential therapeutic agents. However, studies in children are much more limited. Here we review current knowledge pertaining to circulating progenitor cells in the context of childhood cardiovascular disease. Priorities for further research are also highlighted.
Collapse
Affiliation(s)
- Eric J Kort
- DeVos Cardiovascular Research Program of Spectrum Health and Van Andel Research Institute, 100 Michigan Street NE, Grand Rapids, MI, 49503, USA.
- Michigan State University, College of Human Medicine, 15 Michigan Street NE, Grand Rapids, MI, USA.
- Helen DeVos Children's Hospital, 100 Michigan Street NE, Grand Rapids, MI, USA.
| | - Lacey Croskey
- Michigan State University, College of Human Medicine, 15 Michigan Street NE, Grand Rapids, MI, USA
| | - Taryn Scibienski
- Michigan State University, College of Human Medicine, 15 Michigan Street NE, Grand Rapids, MI, USA
| | - Surender Rajasekaran
- Michigan State University, College of Human Medicine, 15 Michigan Street NE, Grand Rapids, MI, USA
- Helen DeVos Children's Hospital, 100 Michigan Street NE, Grand Rapids, MI, USA
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program of Spectrum Health and Van Andel Research Institute, 100 Michigan Street NE, Grand Rapids, MI, 49503, USA
- Michigan State University, College of Human Medicine, 15 Michigan Street NE, Grand Rapids, MI, USA
- Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
127
|
Bartaula-Brevik S, Pedersen TO, Finne-Wistrand A, Bolstad AI, Mustafa K. Angiogenic and Immunomodulatory Properties of Endothelial and Mesenchymal Stem Cells. Tissue Eng Part A 2016; 22:244-52. [PMID: 26650611 PMCID: PMC4779276 DOI: 10.1089/ten.tea.2015.0316] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/17/2015] [Indexed: 12/12/2022] Open
Abstract
It has been suggested that the effect of implanted cells on the local environment is important when selecting the appropriate cell type for tissue regeneration. Our aim was to compare the local tissue response to implanted human mesenchymal stem cells (MSC) and human umbilical vein endothelial cells (EC). MSC and EC were cultured in poly(L-lactide-co-1,5-dioxepan-2-one) scaffolds for 1 week in a bioreactor system, after which they were implanted subcutaneously in NOD/SCID mice. After 3 weeks, scaffolds were retrieved, and the mRNA expression of selected genes involved in hypoxia and inflammation was examined by real-time reverse transcription polymerase chain reaction and correlated with immunofluorescent staining for corresponding proteins. The Toll-like receptor signaling pathway was examined by superarray hybridization. The expression of 53 angiogenesis-related proteins was investigated by a proteome profiler angiogenesis antibody array kit. Vascularization was quantified using immunohistochemistry for CD31. The expression of hypoxia-inducible factors and biomarkers for angiogenesis was more strongly upregulated in response to implanted EC than to MSC, suggesting a higher sensitivity to low oxygen tension among EC. Hypoxic signaling was increased after implantation of EC compared with MSC, leading to a prolonged acute inflammatory phase that promoted ingrowth of vascular cells and establishment of the circulation. Inflammatory cytokines were also differently expressed at the gene and protein levels in the two experimental groups, resulting in altered recruitment of acute and chronic inflammatory cells. The end result of these differences was increased vessel formation within the constructs in the EC group.
Collapse
Affiliation(s)
| | | | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Kamal Mustafa
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| |
Collapse
|
128
|
Mount S, Davis DR. Electrical effects of stem cell transplantation for ischaemic cardiomyopathy: friend or foe? J Physiol 2016; 594:2511-24. [PMID: 26584682 DOI: 10.1113/jp270540] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/18/2015] [Indexed: 01/07/2023] Open
Abstract
Despite advances in other realms of cardiac care, the mortality attributable to ischaemic cardiomyopathy has only marginally decreased over the last 10 years. These findings highlight the growing realization that current pharmacological and device therapies rarely reverse disease progression and rationalize a focus on novel means to reverse, repair and re-vascularize damaged hearts. As such, multiple candidate cell types have been used to regenerate damaged hearts either directly (through differentiation to form new tissue) or indirectly (via paracrine effects). Emerging literature suggests that robust engraftment of electrophysiolgically heterogeneous tissue from transplanted cells comes at the cost of a high incidence of ventricular arrhythmias. Similar electrophysiological studies of haematological stem cells raised early concerns that transplant of depolarized, inexcitable cells that also induce paracrine-mediated electrophysiological remodelling may be pro-arrhythmic. However, meta-analyses suggest that patients receiving haematological stem cells paradoxically may experience a decrease in ventricular arrhythmias, an observation potentially related to the extremely poor long-term survival of injected cells. Finally, early clinical and preclinical data from technologies capable of differentiating to a mature cardiomyocyte phenotype (such as cardiac-derived stem cells) suggests that these cells are not pro-arrhythmic although they too lack robust long-term engraftment. These results highlight the growing understanding that as next generation cell therapies are developed, emphasis should also be placed on understanding possible anti-arrhythmic contributions of transplanted cells while vigilance is needed to predict and treat the inadvertent effects of regenerative cell therapies on the electrophysiological stability of the ischaemic cardiomyopathic heart.
Collapse
Affiliation(s)
- Seth Mount
- University of Ottawa Heart Institute, Ottawa, Canada, K1Y 4W7
| | - Darryl R Davis
- University of Ottawa Heart Institute, Ottawa, Canada, K1Y 4W7
| |
Collapse
|
129
|
Okada K, Kawao N, Yano M, Tamura Y, Kurashimo S, Okumoto K, Kojima K, Kaji H. Stromal cell-derived factor-1 mediates changes of bone marrow stem cells during the bone repair process. Am J Physiol Endocrinol Metab 2016; 310:E15-23. [PMID: 26530150 DOI: 10.1152/ajpendo.00253.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/27/2015] [Indexed: 12/11/2022]
Abstract
Osteoblasts, osteoclasts, chondrocytes, and macrophages that participate in the bone repair process are derived from hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). However, the roles of these stem cells during the repair of injured bone tissue are still unclear. In the present study, we examined the effects of bone defect on HSCs and MSCs in bone marrow and spleen in 75 mice and its mechanism. We analyzed the HSC and MSC populations in these tissues of a mouse with femoral bone damage by using flow cytometry. The number of HSCs in the bone marrow of mice with damaged femurs was significantly lower than the number of these cells in the bone marrow of the contralateral intact femurs on day 2 after injury. Meanwhile, the number of MSCs in the bone marrow of mice with damaged femurs was significantly higher than that of the contralateral femurs. Both intraperitoneal administration of AMD3100, a C-X-C chemokine receptor 4 (CXCR4) antagonist, and local treatment with an anti-stromal cell-derived factor-1 (SDF-1) antibody blunted the observed decrease in HSC and increase in MSC populations within the bone marrow of injured femurs. In conclusion, the present study revealed that there is a concurrent decrease and increase in the numbers of HSCs and MSCs, respectively, in the bone marrow during repair of mouse femoral bone damage. Furthermore, the SDF-1/CXCR4 system was implicated as contributing to the changes in these stem cell populations upon bone injury.
Collapse
Affiliation(s)
- Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Masato Yano
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Yukinori Tamura
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Shinzi Kurashimo
- Life Science Research Institute, Kinki University, Osakasayama, Osaka, Japan
| | - Katsumi Okumoto
- Life Science Research Institute, Kinki University, Osakasayama, Osaka, Japan
| | - Kotarou Kojima
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| |
Collapse
|
130
|
Wang DD, Yang M, Zhu Y, Mao C. Reiterated Targeting Peptides on the Nanoparticle Surface Significantly Promote Targeted Vascular Endothelial Growth Factor Gene Delivery to Stem Cells. Biomacromolecules 2015; 16:3897-903. [PMID: 26588028 PMCID: PMC4922499 DOI: 10.1021/acs.biomac.5b01226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Nonviral gene delivery vectors hold great promise for gene therapy due to the safety concerns with viral vectors. However, the application of nonviral vectors is hindered by their low transfection efficiency. Herein, in order to tackle this challenge, we developed a nonviral vector integrating lipids, sleeping beauty transposon system and 8-mer stem cell targeting peptides for safe and efficient gene delivery to hard-to-transfect mesenchymal stem cells (MSCs). The 8-mer MSC-targeting peptides, when synthetically reiterated in three folds and chemically presented on the surface, significantly promoted the resultant lipid-based nanoparticles (LBNs) to deliver VEGF gene into MSCs with a high transfection efficiency (∼52%) and long-lasting gene expression (for longer than 170 h) when compared to nonreiterated peptides. However, the reiterated stem cell targeting peptides do not enable the highly efficient gene transfer to other control cells. This work suggests that the surface presentation of the reiterated stem cell-targeting peptides on the nonviral vectors is a promising method for improving the efficiency of cell-specific nonviral gene transfection in stem cells.
Collapse
Affiliation(s)
- Dong-Dong Wang
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ye Zhu
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
131
|
Steinbach SK, Husain M. Vascular smooth muscle cell differentiation from human stem/progenitor cells. Methods 2015; 101:85-92. [PMID: 26678794 DOI: 10.1016/j.ymeth.2015.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 01/16/2023] Open
Abstract
Transplantation of vascular smooth muscle cells (VSMCs) is a promising cellular therapy to promote angiogenesis and wound healing. However, VSMCs are derived from diverse embryonic sources which may influence their role in the development of vascular disease and in its therapeutic modulation. Despite progress in understanding the mechanisms of VSMC differentiation, there remains a shortage of robust methods for generating lineage-specific VSMCs from pluripotent and adult stem/progenitor cells in serum-free conditions. Here we describe a method for differentiating pluripotent stem cells, such as embryonic and induced pluripotent stem cells, as well as skin-derived precursors, into lateral plate-derived VSMCs including 'coronary-like' VSMCs and neural crest-derived VSMC, respectively. We believe this approach will have broad applications in modeling origin-specific disease vulnerability and in developing personalized cell-based vascular grafts for regenerative medicine.
Collapse
Affiliation(s)
- Sarah K Steinbach
- McEwen Centre for Regenerative Medicine, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Division of Experimental Therapeutics, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada
| | - Mansoor Husain
- McEwen Centre for Regenerative Medicine, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Division of Experimental Therapeutics, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Departments of Medicine, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Departments of Physiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Departments of Laboratory Medicine & Pathobiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Ted Rogers Centre for Heart Research, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Peter Munk Cardiac Centre, University Health Network, 200 Elizabeth St., Toronto, Ontario M5G-2C4, Canada.
| |
Collapse
|
132
|
Can A, Ulus AT, Cinar O, Topal Celikkan F, Simsek E, Akyol M, Canpolat U, Erturk M, Kara F, Ilhan O. Human Umbilical Cord Mesenchymal Stromal Cell Transplantation in Myocardial Ischemia (HUC-HEART Trial). A Study Protocol of a Phase 1/2, Controlled and Randomized Trial in Combination with Coronary Artery Bypass Grafting. Stem Cell Rev Rep 2015; 11:752-760. [PMID: 26123356 DOI: 10.1007/s12015-015-9601-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs), which may be obtained from the bone marrow, have been studied for more than a decade in the setting of coronary artery disease (CAD). Adipose tissue-derived MSCs have recently come into focus and are being tested in a series of clinical trials. MSC-like cells have also been derived from a variety of sources, including umbilical cord stroma, or HUC-MSCs. The HUC-HEART trail (ClinicalTrials.gov Identifier: NCT02323477) is a phase 1/2, controlled, multicenter, randomized clinical study of the intramyocardial delivery of allogeneic HUC-MSCs in patients with chronic ischemic cardiomyopathy. A total of 79 patients (ages 30-80) with left ventricle ejection fractions ranging between 25 and 45% will be randomized in a 2:1:1 pattern in order to receive an intramyocardial injection of either HUC-MSCs or autologous bone marrow-derived mononuclear cells (BM-MNCs) in combination with coronary arterial bypass grafting (CABG) surgery. The control group of patients will receive no cells and undergo CABG alone. Human HUC-MSCs will be isolated, propagated and banked in accordance with a cGMP protocol, whereas the autologous BM-MNCs will be isolated via aspiration from the iliac crest and subsequently process in a closed-circuit cell purification system shortly before cell transplantation. The cell injections will be implemented in 10 peri-infarct areas. Baseline and post-transplantation outcome measures will be primarily utilized to test both the safety and the efficacy of the administered cells for up to 12 months.
Collapse
Affiliation(s)
- Alp Can
- Department of Histology and Embryology, Laboratory for Stem Cells and Reproductive Cell Biology, Ankara University School of Medicine, Ankara, Turkey,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Park CM, Kim MJ, Kim SM, Park JH, Kim ZH, Choi YS. Umbilical cord mesenchymal stem cell-conditioned media prevent muscle atrophy by suppressing muscle atrophy-related proteins and ROS generation. In Vitro Cell Dev Biol Anim 2015; 52:68-76. [DOI: 10.1007/s11626-015-9948-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/31/2015] [Indexed: 12/11/2022]
|
134
|
Mesenchymal Stem Cells for Cardiac Regenerative Therapy: Optimization of Cell Differentiation Strategy. Stem Cells Int 2015; 2015:524756. [PMID: 26339251 PMCID: PMC4539177 DOI: 10.1155/2015/524756] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/28/2015] [Accepted: 03/11/2015] [Indexed: 01/25/2023] Open
Abstract
With the high mortality rate, coronary heart disease (CHD) has currently become a major life-threatening disease. The main pathological change of myocardial infarction (MI) is the induction of myocardial necrosis in infarction area which finally causes heart failure. Conventional treatments cannot regenerate the functional cell efficiently. Recent researches suggest that mesenchymal stem cells (MSCs) are able to differentiate into multiple lineages, including cardiomyocyte-like cells in vitro and in vivo, and they have been used for the treatment of MI to repair the injured myocardium and improve cardiac function. In this review, we will focus on the recent progress on MSCs derived cardiomyocytes for cardiac regeneration after MI.
Collapse
|
135
|
Zhou LN, Zhang JW, Liu XL, Zhou LH. Co-Graft of Bone Marrow Stromal Cells and Schwann Cells Into Acellular Nerve Scaffold for Sciatic Nerve Regeneration in Rats. J Oral Maxillofac Surg 2015; 73:1651-60. [DOI: 10.1016/j.joms.2015.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/06/2015] [Accepted: 02/06/2015] [Indexed: 01/21/2023]
|
136
|
Effect of decellularized tissue powders on a rat model of acute myocardial infarction. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 56:494-500. [PMID: 26249619 DOI: 10.1016/j.msec.2015.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/18/2015] [Accepted: 07/09/2015] [Indexed: 12/22/2022]
Abstract
Many research groups are currently investigating new treatment modalities for myocardial infarction. Numerous aspects need to be considered for the clinical application of these therapies, such as low cell integration and engraftment rates of cell injection techniques. Decellularized tissues are considered good materials for promoting regeneration of traumatic tissues. The properties of the decellularized tissues are sustained after processing to powder form. In this study, we examined the use of decellularized tissue powder in a rat model of acute myocardial infarction. The decellularized tissue powders, especially liver powder, promoted cell integration and neovascularization both in vitro and in vivo. Decellularized liver powder induced neovascularization in the infarct area, resulting in the suppression of myocardial necrosis. The results of this study suggest that decellularized liver powder has good potential for application as a blood supply material for the treatment of myocardial infarction.
Collapse
|
137
|
Adipose-derived Mesenchymal Stem Cells and Their Reparative Potential in Ischemic Heart Disease. ACTA ACUST UNITED AC 2015; 68:599-611. [DOI: 10.1016/j.rec.2015.02.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/23/2015] [Indexed: 12/21/2022]
|
138
|
Sienkiewicz D, Kulak W, Okurowska-Zawada B, Paszko-Patej G, Kawnik K. Duchenne muscular dystrophy: current cell therapies. Ther Adv Neurol Disord 2015; 8:166-77. [PMID: 26136844 PMCID: PMC4480531 DOI: 10.1177/1756285615586123] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Duchenne muscular dystrophy is a genetically determined X-linked disease and the most common, progressive pediatric muscle disorder. For decades, research has been conducted to find an effective therapy. This review presents current therapeutic methods for Duchenne muscular dystrophy, based on scientific articles in English published mainly in the period 2000 to 2014. We used the PubMed database to identify and review the most important studies. An analysis of contemporary studies of stem cell therapy and the use of granulocyte colony-stimulating factor (G-CSF) in muscular dystrophy was performed.
Collapse
Affiliation(s)
- Dorota Sienkiewicz
- Department of Pediatric Rehabilitation Medical University, Bialystok, Poland
| | - Wojciech Kulak
- Department of Pediatric Rehabilitation, Medical University of Białystok, 15-274 Bialystok, 17 Waszyngtona street, Poland
| | | | | | - Katarzyna Kawnik
- Department of Pediatric Rehabilitation Medical University, Bialystok, Poland
| |
Collapse
|
139
|
Badimon L, Oñate B, Vilahur G. Células madre mesenquimales derivadas de tejido adiposo y su potencial reparador en la enfermedad isquémica coronaria. Rev Esp Cardiol 2015. [DOI: 10.1016/j.recesp.2015.02.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
140
|
Yu Z, Zhang H, Yu M, Ye Q. Analysis of Gene Expression During the Development of Congestive Heart Failure After Myocardial Infarction in Rat Models. Int Heart J 2015; 56:444-9. [PMID: 26104178 DOI: 10.1536/ihj.14-422] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Our study aimed to investigate the gene expression at different myocardial infarction (MI) phases and to understand the development mechanisms of congestive heart failure (CHF) after MI. Dataset GSE1957 including 24 samples of rat left ventricles at 1-day post MI or sham operation and 7-day post MI or sham operation was downloaded from Gene Expression Ominibus. The data were normalized with an affyPLM package and differentially expressed genes (DEGs) were identified with a Linear Models for Microarray Data package. Heat maps of the DEGs were constructed using Cluster 3.0. GO (Gene Ontology) enrichment analysis of the DEGs was performed in Database for Annotation, Visualization, and Integrated Discovery. A protein-protein interaction (PPI) network was constructed by Biomolecular Interaction Network Database and visualized by Cytoscape, and a subnetwork was analyzed using plugin ClusterONE in Cytoscape. A total of 5 DEGs at 1-day post-MI, 5 DEGs at 7-day post-MI, and 7 DEGs between the MI and sham groups at 1-day and 7-day post-MI were identified. For the GO category analysis, DEGs at 1-day post-MI were enriched in response to cytokine stimulus. DEGs at 7-day post-MI were enriched in response to inorganic substance and chemical homeostasis. DEGs between 1-day and 7-day post-MI including CDK2 and CDC20 were significantly enriched in mitosis. CDK2, ANXA1, CDC20, and AQP2 were included in the PPI network, and CDK2 was the only DEG included in the subnetwork. In conclusion, the induction of DEGs at 7-day post-MI might participate in the response to a hormone and endogenous stimulus to regulate the development of CHF after MI.
Collapse
Affiliation(s)
- Zhuo Yu
- Cardiology, 2) Cardiovascular Surgery, the First Affiliated Hospital of Kunming University the Third Hospital of Kunming, Kunming, China
| | | | | | | |
Collapse
|
141
|
Therapeutic angiogenesis promotes efficacy of human umbilical cord matrix stem cell transplantation in cardiac repair. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18. [PMID: 26221480 PMCID: PMC4509951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Although previous studies have confirmed the beneficial effects of human umbilical cord matrix stem cell (hUCM) transplantation post myocardial infarction (MI), but this stem cell resource has no potential to induce angiogenesis. In order to achieve the process of angiogenesis and cardiomyocyte regeneration, two required factors for cardiac repair agents were examined namely; hUCM and VEGF on an infarcted heart. The main objective of this research is to investigate the combinatory effect of dhUCM and VEGF transplantation on an infarcted heart. MATERIALS AND METHODS 45 min of ligating the left anterior descending coronary artery, the MI-induced animals received 50 μl PBS, 5 μg VEGF, 5×10(6) hUCM cells alone, combined with 5 μg VEGF and 5×10(6) differentiated hUCM cells alone or combined with 5 μg VEGF through intramyocardial injection. MI group, without hUCM and VEGF served as the control group. Left ventricular function and angiogenesis were also evaluated. RESULTS After eight weeks post MI, there were significant rise in left ventricular ejection farction in dhUCM+VEGF group compared to the other treated and non-treated groups (P<0.05). Fibrosis tissue was markedly lower in the dhUCM+VEGF and hUCM+VEGF groups compared to the other treated and non-treated groups (P<0.05). Despite these benefits, vascular density in dhUCM+VEGF group was not markedly different compared to VEGF and hUCM+VEGF groups. The transplanted hUCM and dhUCM cells survived and migrated to the infarcted area. CONCLUSION Our findings demonstrated that the dhUCM cells transplantation combined with VEGF were more efficient on an infarcted heart.
Collapse
|
142
|
Chen YL, Sun CK, Tsai TH, Chang LT, Leu S, Zhen YY, Sheu JJ, Chua S, Yeh KH, Lu HI, Chang HW, Lee FY, Yip HK. Adipose-derived mesenchymal stem cells embedded in platelet-rich fibrin scaffolds promote angiogenesis, preserve heart function, and reduce left ventricular remodeling in rat acute myocardial infarction. Am J Transl Res 2015; 7:781-803. [PMID: 26175843 PMCID: PMC4494133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/11/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE This study tested the hypothesis that autologous adipose-derived mesenchymal stem cells (ADMSCs) embedded in platelet-rich fibrin (PRF) can significant promote myocardial regeneration and repair after acute myocardial infarction (AMI). SUMMARY BACKGROUND With avoiding the needle-related complications, PRF-embedded autologous ADMSCs graft provides a new effective stem cell-based therapeutic strategy for myocardial repair. METHODS Adult male Sprague-Dawley rats were equally divided (n = 8 per group) into group 1 (sham-operated), group 2 (AMI by ligating left coronary artery), group 3 (AMI+ PRF), and group 4 (AMI+PRF-embedded autologous ADMSCs). RPF with or without ADMSCs was patched on infarct area 1h after AMI induction. All animals were sacrificed on day 42 after echocardiography. RESULTS Left ventricular (LV) dimension and infarct/fibrotic areas were lowest in group 1, highest in group 2, in group 3 higher than in group 4, whereas LV performance and wall thickness exhibited a reversed pattern in all groups (all p < 0.001). Protein expressions of inflammatory (MMP-9, IL-1β), oxidative, apoptotic (Bax, cleaved PARP), fibrotic (Smad 3, TFG-β), hypertrophic (β-MHC), and heart failure (BNP) biomarkers displayed an identical pattern in infarct/fibrotic areas, whereas the protein expressions of anti-inflammatory (IL-10), anti-apoptotic (Bcl-2), anti-fibrotic (Smad1/5, BMP-2) biomarkers and α-MHC showed an opposite pattern (all p < 0.01). Angiogenic activities (c-Kit+, Sca-1+, CD31+, SDF-1α+, CXCR4+ cells; protein expressions of SDF-1α, CXCR4, VEGF) were highest in group 4 and lowest in group 1 (all p < 0.001). CONCLUSION ADMSCs embedded in PRF offered significant benefit in preserving LV function and limiting LV remodeling after AMI.
Collapse
Affiliation(s)
- Yung-Lung Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International StudentsKaohsiung, 82245, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Li-Teh Chang
- Basic Science, Nursing Department, Meiho Institute of TechnologyPingtung, 91202, Taiwan
| | - Steve Leu
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Yen-Yi Zhen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Jiunn-Jye Sheu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Sarah Chua
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Kuo-Ho Yeh
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Hung-I Lu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Hsueh-Wen Chang
- Department of Biological Sciences, National Sun Yat-Sen UniversityKaohsiung, 80424, Taiwan
| | - Fan-Yen Lee
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung, 83301, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical UniversityTaichung, 40402, Taiwan
| |
Collapse
|
143
|
De Angelis B, Gentile P, Orlandi F, Bocchini I, Di Pasquali C, Agovino A, Gizzi C, Patrizi F, Scioli MG, Orlandi A, Cervelli V. Limb rescue: a new autologous-peripheral blood mononuclear cells technology in critical limb ischemia and chronic ulcers. Tissue Eng Part C Methods 2015; 21:423-435. [PMID: 25341088 DOI: 10.1089/ten.tec.2014.0245] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Therapeutic angiogenesis by autologous-peripheral blood mononuclear cells (A-PBMNC) implantation has been shown to be a safe and effective treatment for critical limb ischemia (CLI). We herein report our investigation of the long-term efficacy of implantation of A-PBMNC produced by selective filtration to treat patients with CLI, for which surgical bypass and/or percutaneous transluminal angioplasty are not possible. MATERIALS AND METHODS This is a prospective, and not a randomized, study based on a treated group who did not respond to conventional therapy (n=43) when implanted with A-PBMNC cells versus a historically matched control group. Patients of both groups were suffering from CLI Fontaine scale IV with chronic ulcers and various accompanying conditions (diabetes, heart disease, kidney failure, etc.). Treated patients were implanted with 12 mL of A-PBMNC, 0.2-0.3 mL for each bolus, collected by selective filtration from 120 mL of peripheral blood in the ischemic area of the limbs. Patients were not mobilized by granulocyte colony-stimulating factor, and the A-PBMNC treatment was repeated for a maximum of three times. RESULTS The A-PBMNC-treated group showed a statistically significant improvement of limb rescue of 95.3% versus 52.2% of the control group (p<0.001), and the result had been maintained for 2 years. The A-PBMNC group also showed reduction in pain at rest, increased maximum walking distance, and healing of the wound, which led to an overall improvement in the quality of life. Post-treatment radiological studies showed an improvement of vascularization with the formation of new collateral and by histological findings. Within 2 years of follow-up, none of the patients whom we treated showed any major or systemic adverse effects. CONCLUSION The local injection of A-PBMNC showed striking early and long-term effects together with a favorable safety profile, significantly decreasing the risk of amputation. Our results are comparable with published data obtained by injection of bone marrow mononuclear cells, but with a lot less invasive approach. Moreover the intraoperative selective filtration system we used is fast, safe, not operator dependent, and easy to use in a sterile operating theatre. This system aims to produce fresh A-PBMNC as a valuable treatment option, particularly for those difficult patients who cannot undergo revascularization.
Collapse
Affiliation(s)
- Barbara De Angelis
- 1 Department of Plastic and Reconstructive Surgery, University of Rome Tor Vergata , Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Zahra SA, Muzavir SR, Ashraf S, Ahmad A. Stem cell research in pakistan; past, present and future. Int J Stem Cells 2015; 8:1-8. [PMID: 26019749 PMCID: PMC4445703 DOI: 10.15283/ijsc.2015.8.1.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 02/02/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Stem cells have proved to have great therapeutic potential as stem cell treatment is replacing traditional ways of treatment in different disorders like cancer, aplastic anemia, stroke, heart disorders. The developed and developing countries are investing differently in this area of research so research output and clinical translation of research greatly vary among developed and developing countries. Present study was done to investigate the current status of stem cells research in Pakistan and ways to improve it. RESULTS Many advanced countries (USA, UK and Canada etc.) are investing heavily in stem cell research and treatment. Different developing countries like Iran, Turkey and India are also following the developed countries and investing a lot in stem cells research. Pakistan is also making efforts in establishing this field to get desired benefits but unfortunately the progress is at very low pace. If Government plays an active role along with private sector, stem cell research in Pakistan can be boosted up. The numbers of publications from Pakistan are very less compared to developed and neighboring countries and Pakistan also has very less number of institutes working in this area of research. CONCLUSIONS Stem cells research is at its initial stages in Pakistan and there is great need to bring Government, academia and industry together so they could make serious efforts to promote research in this very important field. This will help millions of patients suffering from incurable disorders and will also reduce economic loss.
Collapse
Affiliation(s)
- Sayeda Anum Zahra
- National Academy of Young Scientists (NAYS), University of the Punjab, Quaid-i-Azam Campus, Lahore,
Pakistan
| | - Sayed Raheel Muzavir
- National Academy of Young Scientists (NAYS), University of the Punjab, Quaid-i-Azam Campus, Lahore,
Pakistan
| | - Sadia Ashraf
- National Academy of Young Scientists (NAYS), University of the Punjab, Quaid-i-Azam Campus, Lahore,
Pakistan
| | - Aftab Ahmad
- National Academy of Young Scientists (NAYS), University of the Punjab, Quaid-i-Azam Campus, Lahore,
Pakistan
| |
Collapse
|
145
|
Lerman DA, Alotti N, Ume KL, Péault B. Cardiac Repair and Regeneration: The Value of Cell Therapies. Eur Cardiol 2015; 11:43-48. [PMID: 27499812 DOI: 10.15420/ecr.2016:8:1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ischaemic heart disease is the predominant contributor to cardiovascular morbidity and mortality; one million myocardial Infarctions occur per year in the USA, while more than five million patients suffer from chronic heart failure. Recently, heart failure has been singled out as an epidemic and is a staggering clinical and public health problem associated with significant mortality, morbidity and healthcare expenditures, particularly among those aged ≥65 years. Death rates have improved dramatically over the last four decades, but new approaches are nevertheless urgently needed for those patients who go on to develop ventricular dysfunction and chronic heart failure. Over the past decade, stem cell transplantation has emerged as a promising therapeutic strategy for acute or chronic ischaemic cardiomyopathy. Multiple candidate cell types have been used in preclinical animal models and in humans to repair or regenerate the injured heart, either directly or indirectly (through paracrine effects), including: embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), neonatal cardiomyocytes, skeletal myoblasts (SKMs), endothelial progenitor cells, bone marrow mononuclear cells (BMMNCs), mesenchymal stem cells (MSCs) and, most recently, cardiac stem cells (CSCs). Although no consensus has emerged yet, the ideal cell type for the treatment of heart disease should: (a) improve heart function; (b) create healthy and functional cardiac muscle and vasculature, integrated into the host tissue; (c) be amenable to delivery by minimally invasive clinical methods; (d) be available 'off the shelf' as a standardised reagent; (e) be tolerated by the immune system; (f) be safe oncologically, i.e. not create tumours; and (g) circumvent societal ethical concerns. At present, it is not clear whether such a 'perfect' stem cell exists; what is apparent, however, is that some cell types are more promising than others. In this brief review, we provide ongoing data on agreement and controversy arising from clinical trials and touch upon the future directions of cell therapy for heart disease.
Collapse
Affiliation(s)
- Daniel Alejandro Lerman
- Department of Cardiothoracic Surgery, Royal Infirmary Hospital of Edinburgh (NHS Lothian), University of Edinburgh, Scotland, UK; MRC Centre for Regenerative Medicine and College of Medicine and Veterinary, University of Edinburgh, Scotland, UK
| | | | | | - Bruno Péault
- MRC Centre for Regenerative Medicine and College of Medicine and Veterinary, University of Edinburgh, Scotland, UK; David Geffen School of Medicine at UCLA, Orthopaedic Hospital Research Centre, University of California at Los Angeles, USA
| |
Collapse
|
146
|
Endothelial progenitor cells in tumor angiogenesis: another brick in the wall. Stem Cells Int 2015; 2015:832649. [PMID: 26000021 PMCID: PMC4427119 DOI: 10.1155/2015/832649] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 12/14/2022] Open
Abstract
Until 15 years ago, vasculogenesis, the formation of new blood vessels from undifferentiated cells, was thought to occur only during embryonic development. The discovery of circulating cells that are able to promote vascular regeneration and repair—the so-called endothelial progenitor cells (EPCs)—changed that, and EPCs have since been studied extensively. It is already known that EPCs include many subtypes of cells that play a variety of roles in promoting vascular growth. Some EPCs are destined to differentiate into endothelial cells, whereas others are capable of promoting and sustaining angiogenesis through paracrine mechanisms. Vasculogenesis and angiogenesis might constitute complementary mechanisms for postnatal neovascularization, and EPCs could be at the core of this process. Although the formation of new blood vessels from preexisting vasculature plays a beneficial role in many physiological processes, such as wound healing, it also contributes to tumor growth and metastasis. However, many aspects of the role played by EPCs in tumor angiogenesis remain unclear. This review aims to address the main aspects of EPCs differentiation and certain characteristics of their main function, especially in tumor angiogenesis, as well as the potential clinical applications.
Collapse
|
147
|
Tian S, Liu Q, Gnatovskiy L, Ma PX, Wang Z. Heart Regeneration with Embryonic Cardiac Progenitor Cells and Cardiac Tissue Engineering. ACTA ACUST UNITED AC 2015; 1. [PMID: 26744736 DOI: 10.19104/jstb.2015.104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide. Recent advances in stem cell research hold great potential for heart tissue regeneration through stem cell-based therapy. While multiple cell types have been transplanted into MI heart in preclinical studies or clinical trials, reduction of scar tissue and restoration of cardiac function have been modest. Several challenges hamper the development and application of stem cell-based therapy for heart regeneration. Application of cardiac progenitor cells (CPCs) and cardiac tissue engineering for cell therapy has shown great promise to repair damaged heart tissue. This review presents an overview of the current applications of embryonic CPCs and the development of cardiac tissue engineering in regeneration of functional cardiac tissue and reduction of side effects for heart regeneration. We aim to highlight the benefits of the cell therapy by application of CPCs and cardiac tissue engineering during heart regeneration.
Collapse
Affiliation(s)
- Shuo Tian
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Qihai Liu
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Leonid Gnatovskiy
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X Ma
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Center, The University of Michigan, Ann Arbor, MI 48109, USA; Department of Materials Science and Engineering, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
148
|
Abstract
This review article discusses the mechanisms of cardiomyogenesis in the adult heart. They include the re-entry of cardiomyocytes into the cell cycle; dedifferentiation of pre-existing cardiomyocytes, which assume an immature replicating cell phenotype; transdifferentiation of hematopoietic stem cells into cardiomyocytes; and cardiomyocytes derived from activation and lineage specification of resident cardiac stem cells. The recognition of the origin of cardiomyocytes is of critical importance for the development of strategies capable of enhancing the growth response of the myocardium; in fact, cell therapy for the decompensated heart has to be based on the acquisition of this fundamental biological knowledge.
Collapse
Affiliation(s)
- Annarosa Leri
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | - Marcello Rota
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Francesco S Pasqualini
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Polina Goichberg
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Piero Anversa
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
149
|
Marongiu M, Serra MP, Contini A, Sini M, Strom SC, Laconi E, Marongiu F. Rat-derived amniotic epithelial cells differentiate into mature hepatocytes in vivo with no evidence of cell fusion. Stem Cells Dev 2015; 24:1429-35. [PMID: 25647334 PMCID: PMC4486142 DOI: 10.1089/scd.2014.0532] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Amniotic epithelial cells (AEC) derived from human placenta represent a useful and noncontroversial source for liver-based regenerative medicine. Previous studies suggested that human- and rat-derived AEC differentiate into hepatocyte-like cells upon transplantation. In the retrorsine (RS) model of liver repopulation, clusters of donor-derived cells engrafted in the recipient liver and, importantly, showed characteristics of mature hepatocytes. The aim of the current study was to investigate the possible involvement of cell fusion in the emergence of hepatocyte clusters displaying a donor-specific phenotype. To this end, 4-week-old GFP(+)/DPP-IV(-) rats were treated with RS and then transplanted with undifferentiated AEC isolated from the placenta of DPP-IV(+) pregnant rats at 16-19 days of gestational age. Results indicated that clusters of donor-derived cells were dipeptidyl peptidase type IV (DPP-IV) positive, but did not express the green fluorescent protein (GFP), suggesting that rat amniotic epithelial cells (rAEC) did not fuse within the host parenchyma, as no colocalization of the two tags was observed. Moreover, rAEC-derived clusters expressed markers of mature hepatocytes (eg, albumin, cytochrome P450), but were negative for the expression of biliary/progenitor markers (eg, epithelial cell adhesion molecule [EpCAM]) and did not express the marker of preneoplastic hepatic nodules glutathione S-transferase P (GST-P). These results extend our previous findings on the potential of AEC to differentiate into mature hepatocytes and suggest that this process can occur in the absence of cell fusion with host-derived cells. These studies support the hypothesis that amnion-derived epithelial cells can be an effective cell source for the correction of liver disease.
Collapse
Affiliation(s)
- Michela Marongiu
- 1Experimental Medicine Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Paola Serra
- 1Experimental Medicine Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Antonella Contini
- 1Experimental Medicine Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Marcella Sini
- 1Experimental Medicine Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Stephen C Strom
- 2Division of Pathology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ezio Laconi
- 1Experimental Medicine Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Fabio Marongiu
- 1Experimental Medicine Unit, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
150
|
On the existence of cardiomesenchymal stem cells. Med Hypotheses 2015; 84:511-5. [PMID: 25769705 DOI: 10.1016/j.mehy.2015.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/20/2015] [Indexed: 11/21/2022]
Abstract
The most efficient cells for cardiac regeneration are myocardium-resident cardiac stem cells. However, the limited availability of these cells restricts their utility for cardiac cellular therapy. Mesenchymal stem cells can differentiate into a wide variety of tissues, but it is not simple to accurately direct cell differentiation into a specific lineage, such as cardiac tissue; this renders a low efficiency for cardiac regeneration therapy. Given the heterogeneity of mesenchymal stem cells, it may be possible to find specific stem cell subpopulations with a definite differentiation capacity toward cardiac lineage. A parameter to assess cardiac differentiation specificity could be surface marker expression; a population with an immunophenotype similar to cardiac stem cells may have a superior therapeutic value than unsorted mesenchymal stem cells. We hypothesize the existence of a cell line that combines the expression of cardiac stem cell surface markers with those of mesenchymal stem cells, a suitable name for this population is cardiomesenchymal stem cells (CMSC); such cells would be ideal for cardiac regeneration.
Collapse
|