Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054878

ABSTRACT

Many clinical studies utilizing MSCs (mesenchymal stem cells, mesenchymal stromal cells, or multipotential stromal cells) are underway in multiple clinical settings; however, the ideal approach to prepare these cells in vitro and to deliver them to injury sites in vivo with maximal effectiveness remains a challenge. Here, pretreating MSCs with agents that block the apoptotic pathways were compared with untreated MSCs. The treatment effects were evaluated in the myocardial infarct setting following direct injection, and physiological parameters were examined at 4 weeks post-infarct in a rat permanent ligation model. The prosurvival treated MSCs were detected in the hearts in greater abundance at 1 week and 4 weeks than the untreated MSCs. The untreated MSCs improved ejection fraction in infarcted hearts from 61% to 77% and the prosurvival treated MSCs further improved ejection fraction to 83% of normal. The untreated MSCs improved fractional shortening in the infarcted heart from 52% to 68%, and the prosurvival treated MSCs further improved fractional shortening to 77% of normal. Further improvements in survival of the MSC dose seems possible. Thus, pretreating MSCs for improved in vivo survival has implications for MSC-based cardiac therapies and in other indications where improved cell survival may improve effectiveness.


Subject(s)
Heart/physiopathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/physiopathology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Electrocardiography , Green Fluorescent Proteins/metabolism , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/toxicity , Male , Mesenchymal Stem Cells/drug effects , Myocardial Infarction/pathology , Rats, Inbred Lew , Recovery of Function/drug effects
2.
NPJ Regen Med ; 4: 22, 2019.
Article in English | MEDLINE | ID: mdl-31815001

ABSTRACT

The terms MSC and MSCs have become the preferred acronym to describe a cell and a cell population of multipotential stem/progenitor cells commonly referred to as mesenchymal stem cells, multipotential stromal cells, mesenchymal stromal cells, and mesenchymal progenitor cells. The MSCs can differentiate to important lineages under defined conditions in vitro and in limited situations after implantation in vivo. MSCs were isolated and described about 30 years ago and now there are over 55,000 publications on MSCs readily available. Here, we have focused on human MSCs whenever possible. The MSCs have broad anti-inflammatory and immune-modulatory properties. At present, these provide the greatest focus of human MSCs in clinical testing; however, the properties of cultured MSCs in vitro suggest they can have broader applications. The medical utility of MSCs continues to be investigated in over 950 clinical trials. There has been much progress in understanding MSCs over the years, and there is a strong foundation for future scientific research and clinical applications, but also some important questions remain to be answered. Developing further methods to understand and unlock MSC potential through intracellular and intercellular signaling, biomedical engineering, delivery methods and patient selection should all provide substantial advancements in the coming years and greater clinical opportunities. The expansive and growing field of MSC research is teaching us basic human cell biology as well as how to use this type of cell for cellular therapy in a variety of clinical settings, and while much promise is evident, careful new work is still needed.

3.
Stem Cells ; 35(4): 851-858, 2017 04.
Article in English | MEDLINE | ID: mdl-28294454

ABSTRACT

Mesenchymal stem cell transplantation is undergoing extensive evaluation as a cellular therapy in human clinical trials. Because MSCs are easily isolated and amenable to culture expansion in vitro there is a natural desire to test MSCs in many diverse clinical indications. This is exemplified by the rapidly expanding literature base that includes many in vivo animal models. More recently, MSC-derived extracellular vesicles (EVs), which include exosomes and microvesicles (MV), are being examined for their role in MSC-based cellular therapy. These vesicles are involved in cell-to-cell communication, cell signaling, and altering cell or tissue metabolism at short or long distances in the body. The exosomes and MVs can influence tissue responses to injury, infection, and disease. MSC-derived exosomes have a content that includes cytokines and growth factors, signaling lipids, mRNAs, and regulatory miRNAs. To the extent that MSC exosomes can be used for cell-free regenerative medicine, much will depend on the quality, reproducibility, and potency of their production, in the same manner that these parameters dictate the development of cell-based MSC therapies. However, the MSC exosome's contents are not static, but rather a product of the MSC tissue origin, its activities and the immediate intercellular neighbors of the MSCs. As such, the exosome content produced by MSCs appears to be altered when MSCs are cultured with tumor cells or in the in vivo tumor microenvironment. Therefore, careful attention to detail in producing MSC exosomes may provide a new therapeutic paradigm for cell-free MSC-based therapies with decreased risk. Stem Cells 2017;35:851-858.


Subject(s)
Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Humans , Mesenchymal Stem Cells/cytology , Paracrine Communication , Stem Cell Niche , Wound Healing
4.
Transfus Med Rev ; 30(1): 37-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26689863

ABSTRACT

In the last 10 years, the role of mesenchymal stromal cells (MSCs) in modulating inflammatory and immune responses has been characterized using both in vitro studies and in vivo models of immune disorders. Mesenchymal stromal cell immunomodulatory properties have been linked to various paracrine factors which expression varies depending on the pathologic condition to which the MSCs are exposed. These factors may directly impact key cells of the adaptive immune system, such as T cells. Indeed, coculturing MSCs with T cells in a mixed lymphocyte reaction assay inhibits T-cell proliferation through the secretion of immunomodulatory cytokines. However, in a context of inflammation, MSCs may secrete paracrine factors that influence other immune cell subpopulations such as dendritic cells and macrophages and polarize them toward a tolerogenic phenotype. In vivo, these same immunomodulatory factors are shown to be increased in the serum of animal models presenting with inflammatory diseases treated with MSC administration. In light of the results from these landmark studies, we review the main MSC secreted factors identified to play a role in modulating inflammatory immune responses either in vitro or in vivo, and we assess the impact of these factors on the therapeutic applications of MSC-based cell therapies in immune diseases.


Subject(s)
Immunomodulation/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Paracrine Communication/immunology , Animals , Cell Proliferation , Cytokines/physiology , Dinoprostone/physiology , Humans , Mesenchymal Stem Cells/immunology , T-Lymphocytes/physiology , Transforming Growth Factor beta/physiology
6.
Cells Tissues Organs ; 199(2-3): 140-9, 2014.
Article in English | MEDLINE | ID: mdl-25402318

ABSTRACT

Mesenchymal stem cells (MSCs) are currently undergoing testing in several clinical settings. The propagation of MSCs from multiple species in culture is an important step in furthering our understanding of these progenitor cells. Pim-1, a proto-oncogenic serine/threonine kinase, regulates cell proliferation, survival, and differentiation. Although it has been shown that Pim-1 participates in signal transduction mediating mitogenic action in MSCs, its roles in the modulation of MSC propagation remain to be defined. Understanding of ovine MSCs transduced with Pim-1 may provide improved ovine models for cellular therapy development. Using genetically modified ovine MSCs that constitutively overexpressed Pim-1 (MSC expressing PIM-1 and ZsGreen protein), we evaluated the impact of elevated Pim-1 activity on the proliferation, survival, and differentiation of MSCs in culture. Our results showed that Pim-1 enhanced the intrinsic molecular signals of growth and survival implicated in the mediation of serum signaling under normal culture conditions (10% serum). We found that Pim-1 promoted MSC proliferation irrespectively of the serum concentration, but with a decreased proliferation rate compared to increased serum concentrations, relative to the control vector-transduced MSC expressing ZsGreen protein. Further, Pim-1 prevented MSC apoptosis induced by hypoxia or serum deprivation as evidenced by enhanced mitochondria integrity and reduced annexin V binding. Interestingly, the phenotype and multilineage differentiation potential of the cells were not influenced by Pim-1. Taken together, these observations demonstrate that Pim-1 kinase cooperates with exogenous serum signals supporting MSC propagation in the ovine model.


Subject(s)
Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , Flow Cytometry , Mesenchymal Stem Cells/cytology , Proto-Oncogene Proteins c-pim-1/genetics , Sheep
8.
Stem Cells Transl Med ; 1(9): 685-95, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23197875

ABSTRACT

Progressive cardiac remodeling, including the myopathic process in the adjacent zone following myocardial infarction (MI), contributes greatly to the development of cardiac failure. Cardiomyoplasty using bone marrow-derived mesenchymal stem cells (MSCs) has been demonstrated to protect cardiomyocytes and/or repair damaged myocardium, leading to improved cardiac performance, but the therapeutic effects on cardiac remodeling are still under investigation. Here, we tested the hypothesis that MSCs could improve the pathological remodeling of the adjacent myocardium abutting the infarct. Allogeneic ovine MSCs were transplanted into the adjacent zone by intracardiac injection 4 hours after infarction. Results showed that remodeling and contractile strain alteration were reduced in the adjacent zone of the MSC-treated group. Cardiomyocyte hypertrophy was significantly attenuated with the normalization of the hypertrophy-related signaling proteins phosphatidylinositol 3-kinase α (PI3Kα), PI3Kγ, extracellular signal-regulated kinase (ERK), and phosphorylated ERK (p-ERK) in the adjacent zone of the MSC-treated group versus the MI-alone group. Moreover, the imbalance of the calcium-handling proteins sarcoplasmic reticulum Ca(2+) adenosine triphosphatase (SERCA2a), phospholamban (PLB), and sodium/calcium exchanger type 1 (NCX-1) induced by MI was prevented by MSC transplantation, and more strikingly, the activity of SERCA2a and uptake of calcium were improved. In addition, the upregulation of the proapoptotic protein Bcl-xL/Bcl-2-associated death promoter (BAD) was normalized, as was phospho-Akt expression; there was less fibrosis, as revealed by staining for collagen; and the apoptosis of cardiomyocytes was significantly inhibited in the adjacent zone by MSC transplantation. Collectively, these data demonstrate that MSC implantation improved the remodeling in the region adjacent to the infarct after cardiac infarction in the ovine infarction model.


Subject(s)
Mesenchymal Stem Cell Transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Ventricular Remodeling , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Class II Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Mesenchymal Stem Cells , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Proto-Oncogene Proteins c-akt/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sheep , Sodium-Calcium Exchanger/metabolism , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/metabolism
10.
Methods Mol Biol ; 449: 27-44, 2008.
Article in English | MEDLINE | ID: mdl-18370081

ABSTRACT

Mesenchymal stem cells (MSCs), sometimes referred to as marrow stromal cells or multipotential stromal cells, represent a class of adult progenitor cells capable of differentiation to several mesenchymal lineages. They can be isolated from many tissues although bone marrow has been used most often. The MSCs may prove useful for repair and regeneration of a variety of mesenchymal tissues such as bone, cartilage, muscle, marrow stroma, and the cells produce useful growth factors and cytokines that may help repair additional tissues. There is also evidence for their differentiation to nonmesenchymal lineages, but that work will not be considered here. This chapter will provide the researcher with some background, and then provide details on MSC isolation, expansion and multilineage differentiation. These are the beginning steps toward formulating tissue repair strategies. The methods provided here have been used in many laboratories around the world and the reader can begin by following the methods presented here, and then test other methods if these prove unsatisfactory for your intended purpose.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Adult , Cell Differentiation , Cell Lineage , Cells, Cultured , Centrifugation, Density Gradient , Humans
11.
Stroke ; 39(5): 1569-74, 2008 May.
Article in English | MEDLINE | ID: mdl-18323495

ABSTRACT

BACKGROUND AND PURPOSE: In animal models of stroke, functional improvement has been obtained after stem cell transplantation. Successful therapy depends largely on achieving a robust and targeted cell engraftment, with intraarterial (IA) injection being a potentially attractive route of administration. We assessed the suitability of laser Doppler flow (LDF) signal measurements and magnetic resonance (MR) imaging for noninvasive dual monitoring of targeted IA cell delivery. METHODS: Transient cerebral ischemia was induced in adult Wistar rats (n=25) followed by IA or intravenous (IV) injection of mesenchymal stem cells (MSCs) labeled with superparamagnetic iron oxide. Cell infusion was monitored in real time with transcranial laser Doppler flowmetry while cellular delivery was assessed with MRI in vivo (4.7 T) and ex vivo (9.4 T). RESULTS: Successful delivery of magnetically labeled MSCs could be readily visualized with MRI after IA but not IV injection. IA stem cell injection during acute stroke resulted in a high variability of cerebral engraftment. The amount of LDF reduction during cell infusion (up to 80%) was found to correlate well with the degree of intracerebral engraftment, with low LDF values being associated with significant morbidity. CONCLUSIONS: High cerebral engraftment rates are associated with impeded cerebral blood flow. Noninvasive dual-modality imaging enables monitoring of targeted cell delivery, and through interactive adjustment may improve the safety and efficacy of stem cell therapy.


Subject(s)
Brain Ischemia/therapy , Cerebral Arteries/surgery , Ischemic Attack, Transient/therapy , Laser-Doppler Flowmetry/methods , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation/methods , Animals , Cell Movement/physiology , Cell Survival/physiology , Cells, Cultured , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cerebrovascular Circulation/physiology , Female , Graft Survival/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Monitoring, Intraoperative/methods , Rats , Rats, Inbred F344 , Rats, Wistar , Stroke/therapy , Treatment Outcome
12.
Circulation ; 112(10): 1451-61, 2005 Sep 06.
Article in English | MEDLINE | ID: mdl-16129797

ABSTRACT

BACKGROUND: Recent results from animal studies suggest that stem cells may be able to home to sites of myocardial injury to assist in tissue regeneration. However, the histological interpretation of postmortem tissue, on which many of these studies are based, has recently been widely debated. METHODS AND RESULTS: With the use of the high sensitivity of a combined single-photon emission CT (SPECT)/CT scanner, the in vivo trafficking of allogeneic mesenchymal stem cells (MSCs) colabeled with a radiotracer and MR contrast agent to acute myocardial infarction was dynamically determined. Redistribution of the labeled MSCs after intravenous injection from initial localization in the lungs to nontarget organs such as the liver, kidney, and spleen was observed within 24 to 48 hours after injection. Focal and diffuse uptake of MSCs in the infarcted myocardium was already visible in SPECT/CT images in the first 24 hours after injection and persisted until 7 days after injection and was validated by tissue counts of radioactivity. In contrast, MRI was unable to demonstrate targeted cardiac localization of MSCs in part because of the lower sensitivity of MRI. CONCLUSIONS: Noninvasive radionuclide imaging is well suited to dynamically track the biodistribution and trafficking of mesenchymal stem cells to both target and nontarget organs.


Subject(s)
Mesenchymal Stem Cell Transplantation , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/therapy , Stem Cells/diagnostic imaging , Tomography, Emission-Computed, Single-Photon/methods , Animals , Cell Differentiation , Cell Division , Cell Survival , Dogs , Indium Radioisotopes , Injections, Intravenous , Magnetic Resonance Imaging , Mesenchymal Stem Cell Transplantation/adverse effects , Organometallic Compounds , Oxyquinoline/analogs & derivatives , Reproducibility of Results , Tomography, Emission-Computed, Single-Photon/standards , Tomography, X-Ray Computed , Transplantation, Homologous
14.
Blood ; 105(4): 1815-22, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15494428

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells found in several adult tissues. Transplanted allogeneic MSCs can be detected in recipients at extended time points, indicating a lack of immune recognition and clearance. As well, a role for bone marrow-derived MSCs in reducing the incidence and severity of graft-versus-host disease (GVHD) during allogeneic transplantation has recently been reported; however, the mechanisms remain to be investigated. We examined the immunomodulatory functions of human MSCs (hMSCs) by coculturing them with purified subpopulations of immune cells and report here that hMSCs altered the cytokine secretion profile of dendritic cells (DCs), naive and effector T cells (T helper 1 [T(H)1] and T(H)2), and natural killer (NK) cells to induce a more anti-inflammatory or tolerant phenotype. Specifically, the hMSCs caused mature DCs type 1 (DC1) to decrease tumor necrosis factor alpha (TNF-alpha) secretion and mature DC2 to increase interleukin-10 (IL-10) secretion; hMSCs caused T(H)1 cells to decrease interferon gamma (IFN-gamma) and caused the T(H)2 cells to increase secretion of IL-4; hMSCs caused an increase in the proportion of regulatory T cells (T(Regs)) present; and hMSCs decreased secretion of IFN-gamma from the NK cells. Mechanistically, the hMSCs produced elevated prostaglandin E2 (PGE(2)) in co-cultures, and inhibitors of PGE(2) production mitigated hMSC-mediated immune modulation. These data offer insight into the interactions between allogeneic MSCs and immune cells and provide mechanisms likely involved with the in vivo MSC-mediated induction of tolerance that could be therapeutic for reduction of GVHD, rejection, and modulation of inflammation.


Subject(s)
Cell Communication/immunology , Mesoderm/cytology , Mesoderm/immunology , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dinoprostone/physiology , Humans , Immunologic Factors/physiology , Immunosuppression Therapy , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Mesoderm/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
16.
NMR Biomed ; 17(7): 513-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15526348

ABSTRACT

Magnetic resonance (MR) tracking of superparamagnetic iron oxide (SPIO)-labeled cells is a relatively new technique to non-invasively determine the biodistribution and migration of transplanted stem cells. A number of studies have recently reported encouraging results in the use of bone marrow-derived mesenchymal stem cells (MSCs) for repair of a variety of tissues. For MR tracking of SPIO-labeled MSCs, it is important to determine the effect that the magnetic labeling procedure may have on the differentiation capacity of labeled MSCs. Human MSCs were labeled with poly-L-lysine (PLL)-coated Feridex, with Feridex being an FDA-approved SPIO formulation in an off-label application, and assayed for cellular differentiation using five different assays. As compared with unlabeled controls, labeled MSCs exhibited an unaltered viability, proliferated similarly, and underwent normal adipogenic and osteogenic differentiation. However, there was a marked inhibition of chondrogenesis. The blocking of chondrogenic activity was mediated by the Feridex, rather than by the transfection agent (PLL). This is the first report showing Feridex blocking of cellular differentiation down a specific pathway (while not affecting viability and proliferation), and caution should thus be exercised when using Feridex-labeled MSCs for chondrogenic MR tracking studies. On the other hand, no detrimental effects of Feridex-labeling are anticipated for MR-guided osteogenic or adipogenic transplantation studies.


Subject(s)
Adipocytes/drug effects , Chondrogenesis/drug effects , Iron/adverse effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Osteogenesis/drug effects , Oxides/adverse effects , Adipocytes/pathology , Cell Differentiation/drug effects , Cells, Cultured , Contrast Media/adverse effects , Dextrans , Dose-Response Relationship, Drug , Ferrosoferric Oxide , Humans , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Staining and Labeling/methods
17.
Circ Res ; 95(1): 9-20, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15242981

ABSTRACT

Mesenchymal stem cells (MSCs) represent a stem cell population present in adult tissues that can be isolated, expanded in culture, and characterized in vitro and in vivo. MSCs differentiate readily into chondrocytes, adipocytes, osteocytes, and they can support hematopoietic stem cells or embryonic stem cells in culture. Evidence suggests MSCs can also express phenotypic characteristics of endothelial, neural, smooth muscle, skeletal myoblasts, and cardiac myocyte cells. When introduced into the infarcted heart, MSCs prevent deleterious remodeling and improve recovery, although further understanding of MSC differentiation in the cardiac scar tissue is still needed. MSCs have been injected directly into the infarct, or they have been administered intravenously and seen to home to the site of injury. Examination of the interaction of allogeneic MSCs with cells of the immune system indicates little rejection by T cells. Persistence of allogeneic MSCs in vivo suggests their potential "off the shelf" therapeutic use for multiple recipients. Clinical use of cultured human MSCs (hMSCs) has begun for cancer patients, and recipients have received autologous or allogeneic MSCs. Research continues to support the desirable traits of MSCs for development of cellular therapeutics for many tissues, including the cardiovascular system. In summary, hMSCs isolated from adult bone marrow provide an excellent model for development of stem cell therapeutics, and their potential use in the cardiovascular system is currently under investigation in the laboratory and clinical settings.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Animals , Bone Marrow Cells/cytology , Cardiomyoplasty/methods , Humans , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/physiology , Mice , Myocardial Infarction/pathology , Neoplasms/therapy , Neovascularization, Physiologic , Rats , Transplantation, Homologous
18.
Circulation ; 107(18): 2290-3, 2003 May 13.
Article in English | MEDLINE | ID: mdl-12732608

ABSTRACT

BACKGROUND: We investigated the potential of magnetic resonance imaging (MRI) to track magnetically labeled mesenchymal stem cells (MR-MSCs) in a swine myocardial infarction (MI) model. METHODS AND RESULTS: Adult farm pigs (n=5) were subjected to closed-chest experimental MI. MR-MSCs (2.8 to 16x107 cells) were injected intramyocardially under x-ray fluoroscopy. MRIs were obtained on a 1.5T MR scanner to demonstrate the location of the MR-MSCs and were correlated with histology. Contrast-enhanced MRI demonstrated successful injection in the infarct and serial MSC tracking was demonstrated in two animals. CONCLUSIONS: MRI tracking of MSCs is feasible and represents a preferred method for studying the engraftment of MSCs in MI.


Subject(s)
Magnetic Resonance Imaging , Mesoderm/cytology , Myocardial Infarction , Myocardium/cytology , Stem Cell Transplantation , Animals , Injections , Iron/analysis , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Stem Cells/chemistry , Swine
20.
Ann Thorac Surg ; 73(6): 1919-25; discussion 1926, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12078791

ABSTRACT

BACKGROUND: A novel therapeutic option for the treatment of acute myocardial infarction involves the use of mesenchymal stem cells (MSCs). The purpose of this study was to investigate whether implantation of autologous MSCs results in sustained engraftment, myogenic differentiation, and improved cardiac function in a swine myocardial infarct model. METHODS: MSCs were isolated and expanded from bone marrow aspirates of 14 domestic swine. A 60-minute left anterior descending artery occlusion was used to produce anterior wall infarction. Piezoelectric crystals were placed within the ischemic region for measurement of regional wall thickness and contractile function. Two weeks later animals autologous, Di-I-labeled MSCs (6 x 10(7)) were implanted into the infarct by direct injection. Hemodynamic and functional measurements were obtained weekly until the time of sacrifice. Immunohistochemistry was used to assess MSC engraftment and myogenic differentiation. RESULTS: Microscopic analysis showed robust engraftment of MSCs in all treated animals. Expression of muscle-specific proteins was seen as early as 2 weeks and could be identified in all animals at sacrifice. The degree of contractile dysfunction was significantly attenuated at 4 weeks in animals implanted with MSCs (5.4% +/- 2.2% versus -3.37% +/- 2.7% in control). In addition, the extent of wall thinning after myocardial infarction was markedly reduced in treated animals. CONCLUSIONS: Mesenchymal stem cells are capable of engraftment in host myocardium, demonstrate expression of muscle specific proteins, and may attenuate contractile dysfunction and pathologic thinning in this model of left ventricular wall infarction. MSC cardiomyoplasty may have significant clinical potential in attenuating the pathology associated with myocardial infarction.


Subject(s)
Disease Models, Animal , Hematopoietic Stem Cell Transplantation/methods , Mesoderm/cytology , Myocardial Infarction/therapy , Animals , Female , Hemodynamics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...