Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(9): e0221457, 2019.
Article in English | MEDLINE | ID: mdl-31479463

ABSTRACT

Freshly isolated, uncultured, autologous adipose derived regenerative cells (ADRCs) have emerged as a promising tool for regenerative cell therapy. The Transpose RT system (InGeneron, Inc., Houston, TX, USA) is a system for isolating ADRCs from adipose tissue, commercially available in Europe as a CE-marked medical device and under clinical evaluation in the United States. This system makes use of the proprietary, enzymatic Matrase Reagent for isolating cells. The present study addressed the question whether the use of Matrase Reagent influences cell yield, cell viability, live cell yield, biological characteristics, physiological functions or structural properties of the ADRCs in final cell suspension. Identical samples of subcutaneous adipose tissue from 12 subjects undergoing elective lipoplasty were processed either with or without the use of Matrase Reagent. Then, characteristics of the ADRCs in the respective final cell suspensions were evaluated. Compared to non-enzymatic isolation, enzymatic isolation resulted in approximately twelve times higher mean cell yield (i.e., numbers of viable cells/ml lipoaspirate) and approximately 16 times more colony forming units. Despite these differences, cells isolated from lipoaspirate both with and without the use of Matrase Reagent were independently able to differentiate into cells of all three germ layers. This indicates that biological characteristics, physiological functions or structural properties relevant for the intended use were not altered or induced using Matrase Reagent. A comprehensive literature review demonstrated that isolation of ADRCs from lipoaspirate using the Transpose RT system and the Matrase Reagent results in the highest viable cell yield among published data regarding isolation of ADRCs from lipoaspirate.


Subject(s)
Cell Separation/methods , Mesenchymal Stem Cells/cytology , Stem Cells/cytology , Subcutaneous Fat/cytology , Adult , Cell Count , Cell Differentiation , Cell Survival , Cell- and Tissue-Based Therapy , Enzymes , Female , Gene Expression , Humans , Indicators and Reagents , Lipectomy , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Middle Aged , Regenerative Medicine , Stem Cells/physiology
2.
Future Sci OA ; 5(5): FSO376, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31245040

ABSTRACT

AIM: Endothelial cells (ECs), isolated from peripheral blood (PB), bone marrow (BM) and cord blood (CB), are limited in numbers and expansion has had limited success. We used a novel serum-free medium (EndoGo) to evaluate effects on ex vivo expansion of CB-derived ECs. MATERIALS & METHODS: Flow cytometry and matrigel were used to determine expansion of ECs and for determination of the EC progenitor cell. RESULTS: EndoGo™-containing cultures demonstrated superior expansion and stimulated proliferation of two distinct subpopulations, CD34+CD31+ and CD34-CD31+, which exhibited different morphology, phenotype and function. EndoGo also expanded the CB endothelial progenitor cells from freshly isolated CB. CONCLUSION: These findings demonstrate the potential of EndoGo to expand CB ECs, which could generate increased numbers of ECs for therapeutic applications.

3.
JCI Insight ; 4(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30944260

ABSTRACT

Multiple myeloma (MM) is an incurable plasma cell malignancy with frequent treatment failures and relapses, suggesting the existence of pathogenic myeloma stem/progenitor populations. However, the identity of MM stem cells remains elusive. We used a murine model of MM with transgenic overexpression of the unfolded protein response sensor X-box binding protein 1 (XBP1s) in the B cell compartment to define MM stem cells. We herein report that a post-germinal center, pre-plasma cell population significantly expands as MM develops. This population has the following characteristics: (a) cell surface phenotype of B220+CD19+IgM-IgD-CD138-CD80+sIgG-AA4.1+FSChi; (b) high expression levels of Pax5 and Bcl6 with intermediate levels of Blimp1 and XBP1s; (c) increased expression of aldehyde dehydrogenase, Notch1, and c-Kit; and (d) ability to efficiently reconstitute antibody-producing capacity in B cell-deficient mice in vivo. We thus have defined a plasma cell progenitor population that resembles myeloma stem cells in mice. These results provide potentially novel insights into MM stem cell biology and may contribute to the development of novel stem cell-targeted therapies for the eradication of MM.


Subject(s)
B-Lymphocytes/pathology , Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , X-Box Binding Protein 1/metabolism , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Humans , Lymphopoiesis , Mice , Mice, Transgenic , Multiple Myeloma/genetics , Primary Cell Culture , Tumor Cells, Cultured , X-Box Binding Protein 1/genetics
4.
Oncotarget ; 9(86): 35611-35622, 2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30479692

ABSTRACT

Incubation of umbilical cord blood (UCB) derived regulatory T-cells (Tregs) with fucosyltransferase enzyme improves their ability to home to the target tissue to prevent graft vs. host disease (GVHD). We report results of 5 patients (Double UCB Transplant, n=2; Peripheral Blood Matched Unrelated Donor Transplant, n=3) who received UCB-Tregs (Dose level = 1×106/kg), infused one day prior to the donor graft. All patients received their designated UCB-Treg dose without any infusion reaction. The ratio of conventional T-cells in donor graft was at least 10 times higher than infused UCB-Tregs (ratio range, 12-356). All patients engrafted at median of 13 days (range, 8-17 days). One patient died due to brain hemorrhage on day 45. A bi-modal increase of plasma IL-10 level occurred on day 7 and day 21 and notably, plasma IL-2 level dropped significantly in all patients at Day 7. All evaluable patients developed ≥grade II acute GVHD and at 1 year follow up, all were alive and without evidence of disease relapse. No increase in the chronic GVHD biomarkers (REG3a and Elafin) was observed at day 7. At the time of last follow up, all evaluable patients were off immune-suppression. Stage 2 of this clinical trial examining UCB-Treg at dose level= 1×107/kg is currently underway.

5.
Oncotarget ; 9(72): 33694-33701, 2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30263095

ABSTRACT

Regulatory T cells (Tregs) are an important component of the immune system involved in regulation of immune cell proliferation and inflammatory responses and preventing autoimmune diseases. The use of Tregs in cellular therapy has recently been explored in clinical trials specifically evaluating the role of ex vivo expanded Tregs in the prevention of graft-versus-host disease during stem cell transplantation. The possibility of Treg use in the clinic requires clinical grade expansion of Tregs for development of cell therapy protocols and proper homing of Tregs to the intended target. Here we demonstrate a novel medium composition to expand CB Tregs, specifically upregulation the homing and activation markers CD62L and cutaneous lymphocyte antigen (CLA). CLA expression was uniquely acquired during activation of Tregs with subsequent loss or lack of expression with media change. This finding highlights the importance of proper growth conditions unique to Tregs that can alter expression of proteins and establishes a baseline for expanding marker specific Tregs that home and target unique tissues.

6.
Methods Mol Biol ; 1441: 203-13, 2016.
Article in English | MEDLINE | ID: mdl-27177668

ABSTRACT

As part of the innate immune system, natural killer (NK) cells are regarded as promising effector cells for adoptive cell therapy approaches to treat patients with cancer. In some cases, genetic modification of the NK cells may be considered but such manipulation has to be integrated into the expansion method to allow the generation of clinically relevant numbers of gene-modified NK cells. Therefore, an efficient gene transfer procedure is needed.Our group developed a retrovirus-based transduction protocol capable of robust expansion of gene-modified NK cells with a high rate of transgene expression. Actively dividing cells is a prerequisite for efficient gene transfer when using a retroviral vector. In the procedure presented here, strong activation of the NK cells was provided by a combination of IL-15 and the K-562 feeder cells. Beside the interest in developing a simple procedure compliant with good manufacturing practice (GMP) for the production of therapeutic products, this approach also provides a valuable means of generating genetically modified primary NK cells for future preclinical studies.


Subject(s)
Interleukin-15/genetics , Killer Cells, Natural/cytology , Transduction, Genetic , Cells, Cultured , Feeder Cells/cytology , Feeder Cells/immunology , Genetic Vectors , Humans , Interleukin-15/metabolism , K562 Cells , Killer Cells, Natural/immunology , Retroviridae/genetics
7.
Leuk Res ; 40: 54-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26601610

ABSTRACT

Leukemic cell lines have become important tools for studies of disease providing a monoclonal cell population that can be extensively expanded in vitro while preserving leukemic cellular characteristics. However, studies of chronic lymphocytic leukemia (CLL) have been impeded in part by the lack of continuous human cell lines. CLL cells have a high spontaneous apoptosis rate in vitro and exhibit minimal proliferation in xenograft models. Therefore, there is a need for development of primary CLL cell lines and we describe the isolation of such a line from the bone marrow of a CLL patient (17p deletion and TP53 mutation) which has been in long term culture for more than 12 months with continuous proliferation. The CLL cell line (termed MDA-BM5) which was generated in vitro with continuous co-culture on autologous stromal cells is CD19+CD5+ and shows an identical pattern of somatic hypermutation as determined in the patient's bone marrow (BM), confirming the origin of the cells from the original CLL clone. MDA-BM5 cells were readily transplantable in NOD/SCID gamma null mice (NSG) with disease developing in the BM, liver and spleen. BM cells from quaternary serial transplantation in NSG mice demonstrated the presence of CD19+CD5+ cells with Ig restricted to lambda which is consistent with the original patient cells. These studies describe a new CLL cell line from a patient with del(17p) that provides a unique model for in vitro and in vivo studies.


Subject(s)
Disease Models, Animal , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Animals , Humans , Mice
8.
Cytotherapy ; 17(11): 1514-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26341480

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs), derived from several tissues including bone marrow and adipose tissue, are being evaluated in clinical trials for a range of diseases. Virtually all tissues of the body contain stromal cells, yet it is unknown whether these sources are similar in phenotype and function. METHODS: We have isolated stromal cells from several human tissues including bone marrow (BM-MSCs), heart (heart stroma, HS), adipose (adipose stroma, AS) and liver (liver stroma, LS) and compared the morphology, phenotype and functional properties of these stromal cell populations. RESULTS: The cellular phenotype of each population was identical, namely, CD105+, CD73+, CD90+, CD34- and CD45-. In addition, morphology and differentiation potential were comparable. Co-culture studies revealed similar supportive potential of BM-MSCs, AS and LS with hematopoietic cells or tumor cells. In contrast, significant inhibition of proliferation of both cells types was obtained with HS, with significant loss of viability with tumor cells, demonstrating a unique functional property of HS with regard to tumor cell proliferation. CONCLUSIONS: Although stromal cells from different tissues have similar morphology and phenotype, their functional properties vary, requiring critical evaluation of stromal cells before use in non-homologous settings. HS may play a key role in inhibiting proliferation of tumor cells in the heart, providing the reason for the low occurrence of tumor development. Given the tumor-supportive property of BM-MSCs and AS, the use of these cells in cardiac tissue may result in replacement of a tumor-inhibitory stroma with a tumor-supportive microenvironment.


Subject(s)
Adipose Tissue/cytology , Bone Marrow Cells/cytology , Liver/cytology , Myocardium/cytology , Stromal Cells/cytology , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Humans , K562 Cells , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phenotype
9.
Cytotherapy ; 17(4): 418-27, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25532425

ABSTRACT

BACKGROUND AIMS: Hematopoietic stem cell transplantation of mobilized peripheral blood progenitor cell (PBPC) products results in rapid platelet engraftment, whereas the use of cord blood (CB) shows significant delays. The difference in the quality and number of megakaryocyte (MK) progenitors that may be responsible for the delay in platelet engraftment has not been fully defined. The objective of this study was to quantify the cells of the MK lineage in PBPC and CB products to determine whether potential differences exist. METHODS: We examined PBPC or CB for differences in surface markers and subpopulations as well as polyploidization status within the MK lineage. Colony-forming assays were used to determine whether differences exist in the clonogenic MK progenitor cell. Finally, we transplanted PBPC and CB mononuclear cells into NOD/SCID/IL2Rγ-/- (NSG) mice to study platelet engraftment rates. RESULTS: Equivalent MK populations and polyploidization was observed in PBPCs and CB. MK progenitors were present only in CD34+ cells and had little difference in colony growth between PBPC and CB. Additionally, MK subpopulations were similar in either product with a slightly more progenitor-enriched phenotype in CB. Finally, when PBPC or CB was transplanted at similar doses, equivalent platelet engraftment rates were observed. CONCLUSIONS: PBPC and CB contain similar frequencies of MK populations, and, when transplanted in comparable doses, CB is as effective as PBPCs in producing platelet engraftment in vivo. Understanding the differences in MK populations between PBPC and CB could help generate protocols to improve platelet engraftment after CB transplantation.


Subject(s)
Blood Platelets/cytology , Leukocytes, Mononuclear/transplantation , Megakaryocyte Progenitor Cells/cytology , Animals , Antigens, CD34/immunology , Cells, Cultured , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Platelet Transfusion
10.
J Hematol Oncol ; 6: 91, 2013 Dec 07.
Article in English | MEDLINE | ID: mdl-24314019

ABSTRACT

Multiple myeloma (MM) is a debilitating disease of proliferating and malignant plasma cells that is currently incurable. The ability of monoclonal recurrence of disease suggests it might arise from a stem cell-like population capable of self-renewal. The difficulty to isolate the cancer stem-like cell in MM has introduced confusion toward this hypothesis. However, recent evidence has suggested that MM originates from the B cell lineage with memory-B cell like features, allowing for self-renewal of the progenitor-like status and differentiation to a monoclonal plasma cell population. Furthermore, this tumor-initiating cell uses signaling pathways and microenvironment similar to the hematopoietic stem cell, though hijacking these mechanisms to create and favor a more tumorigenic environment. The bone marrow niche allows for pertinent evasion, either through avoiding immunosurveillance or through direct interaction with the stroma, inducing quiescence and thus drug resistance. Understanding the interaction of the MM stem cell to the microenvironment and the mechanisms utilized by various stem cell-like populations to allow persistence and therapy-resistance can enable for better targeting of this cell population and potential eradication of the disease.


Subject(s)
Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , Animals , Disease Progression , Humans
11.
Clin Cancer Res ; 19(22): 6242-51, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24077352

ABSTRACT

PURPOSE: gp96 (grp94) is a key downstream chaperone in the endoplasmic reticulum (ER) to mediate unfolded protein response (UPR) and the pathogenesis of multiple myeloma is closely linked to dysregulated UPR. In this study, we aimed to determine the roles of gp96 in the initiation and progression of multiple myeloma in vivo and in vitro. EXPERIMENTAL DESIGN: We generated a mouse model with overexpression of XBP1s and conditional deletion of gp96 in B-cell compartment simultaneously to identify the roles of gp96 in the development of multiple myeloma in vivo. Using a short hairpin RNA (shRNA) system, we silenced gp96 in multiple human multiple myeloma cells and examined the effect of gp96 knockdown on multiple myeloma cells by cell proliferation, cell-cycle analysis, apoptosis assay, immunohistochemistry, and human myeloma xenograft model. The anticancer activity of gp96 selective inhibitor, WS13, was evaluated by apoptosis assay and MTT assay. RESULTS: Genetic deletion of gp96 in XBP1s-Tg mice attenuates multiple myeloma. Silencing of gp96 causes severe compromise in human multiple myeloma cell growth through inhibiting Wnt-LRP-survivin pathway. We also confirmed that knockdown of gp96 decreased human multiple myeloma growth in a murine xenograft model. The targeted gp96 inhibitor induced apoptosis and blocked multiple myeloma cell growth, but did not induce apoptosis in pre-B leukemic cells. We have demonstrated that myeloma growth is dependent on gp96 both genetically and pharmacologically. CONCLUSIONS: gp96 is essential for multiple myeloma cell proliferation and survival, suggesting that gp96 is a novel therapeutic target for multiple myeloma. Clin Cancer Res; 19(22); 6242-51. ©2013 AACR.


Subject(s)
Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Multiple Myeloma/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/biosynthesis , Endoplasmic Reticulum/metabolism , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-1 , Low Density Lipoprotein Receptor-Related Protein-6/biosynthesis , Membrane Glycoproteins/deficiency , Mice , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering , Receptors, LDL/biosynthesis , Regulatory Factor X Transcription Factors , Repressor Proteins/biosynthesis , Survivin , Transcription Factors/biosynthesis , Transplantation, Heterologous , Tumor Suppressor Proteins/biosynthesis , Unfolded Protein Response/drug effects , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/biosynthesis , Wnt Signaling Pathway/drug effects , X-Box Binding Protein 1
12.
Stem Cells ; 31(6): 1202-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23495171

ABSTRACT

The genes and pathways that govern the functions and expansion of hematopoietic stem cells (HSC) are not completely understood. In this study, we investigated the roles of serine/threonine Pim kinases in hematopoiesis in mice. We generated PIM1 transgenic mice (Pim1-Tx) overexpressing human PIM1 driven by vav hematopoietic promoter/regulatory elements. Compared to wild-type littermates, Pim1-Tx mice showed enhanced hematopoiesis as demonstrated by increased numbers of Lin(-) Sca-1 (+) c-Kit (+) (LSK) hematopoietic stem/progenitor cells and cobblestone area forming cells, higher BrdU incorporation in long-term HSC population, and a better ability to reconstitute lethally irradiated mice. We then extended our study using Pim1(-/-), Pim2(-/-), Pim3(-/-) single knockout (KO) mice. HSCs from Pim1(-/-) KO mice showed impaired long-term hematopoietic repopulating capacity in secondary and competitive transplantations. Interestingly, these defects were not observed in HSCs from Pim2(-/-) or Pim3(-/-) KO mice. Limiting dilution competitive transplantation assay estimated that the frequency of LSKCD34(-) HSCs was reduced by approximately 28-fold in Pim1(-/-) KO mice compared to wild-type littermates. Mechanistic studies demonstrated an important role of Pim1 kinase in regulating HSC cell proliferation and survival. Finally, our polymerase chain reaction (PCR) array and confirmatory real-time PCR (RT-PCR) studies identified several genes including Lef-1, Pax5, and Gata1 in HSCs that were affected by Pim1 deletion. Our data provide the first direct evidence for the important role of Pim1 kinase in the regulation of HSCs. Our study also dissects out the relative role of individual Pim kinase in HSC functions and regulation.


Subject(s)
Hematopoietic Stem Cells/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Cell Proliferation , Cell Survival/physiology , Cytokines/metabolism , GATA1 Transcription Factor/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mice, Knockout , Mice, Transgenic/metabolism , Mice, Transgenic/physiology , PAX5 Transcription Factor/metabolism , Receptors, CXCR4/metabolism
13.
Stem Cells Dev ; 20(7): 1143-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21198398

ABSTRACT

Hematopoietic stem cell (HSC) self-renewal is tightly regulated by a complex crosstalk between many cell-intrinsic regulators and a variety of extrinsic signals from the stem cell niche. In this study, we examined whether the p38 mitogen-activated protein kinase (p38) is one of the intrinsic regulators that can negatively regulate HSC self-renewal in vitro and whether inhibition of p38 activity with a small molecule inhibitor can promote HSC expansion ex vivo. The results from this study showed that sorted mouse bone marrow Lin(-)Sca1(+)c-kit(+) cells (LSK(+) cells) exhibited selective activation of p38 after culture in a serum-free medium supplemented with 100 ng/mL stem cell factor, thrombopoietin, and Flt3 ligand. The activation of p38 was associated with a significant reduction in HSCs and induction of apoptosis and cellular senescence in LSK(+) cells and their progeny. Addition of the specific p38 inhibitor SB203580 (SB, 5 µM) to the culture inhibited the activation of p38 in LSK(+) cells, which led to increase in HSC self-renewal and ex vivo expansion as shown by the cobblestone area forming cell assay, competitive repopulation, and serial transplantation. The increase in HSC expansion is likely attributable to SB-mediated inhibition of HSC apoptosis and senescence and upregulation of HoxB4 and CXCR4. These findings suggest that p38 plays an important role in the regulation of HSC self-renewal in vitro and inhibition of p38 activation with a small molecule inhibitor may represent a novel approach to promote ex vivo expansion of HSCs.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/metabolism , Imidazoles/pharmacology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Analysis of Variance , Animals , Apoptosis , Cell Culture Techniques , Cell Differentiation , Cell Growth Processes , Cell Transplantation/methods , Cells, Cultured , Cellular Senescence , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Phosphorylation , Receptors, CXCR4/metabolism , Transcription Factors/metabolism , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Eur Heart J ; 30(22): 2722-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19586959

ABSTRACT

AIMS: The ability of mesenchymal stem cells (MSCs) to heal the chronically injured heart remains controversial. Here we tested the hypothesis that autologous MSCs can be safely injected into a chronic myocardial infarct scar, reduce its size, and improve ventricular function. METHODS AND RESULTS: Female adult Göttingen swine (n = 15) underwent left anterior descending coronary artery balloon occlusion to create reproducible ischaemia-reperfusion infarctions. Bone-marrow-derived MSCs were isolated and expanded from each animal. Twelve weeks post-myocardial infarction (MI), animals were randomized to receive surgical injection of either phosphate buffered saline (placebo, n = 6), 20 million (low dose, n = 3), or 200 million (high dose, n = 6) autologous MSCs in the infarct and border zone. Injections were administered to the beating heart via left anterior thoracotomy. Serial cardiac magnetic resonance imaging was performed to evaluate infarct size, myocardial blood flow (MBF), and left ventricular (LV) function. There was no difference in mortality, post-injection arrhythmias, cardiac enzyme release, or systemic inflammatory markers between groups. Whereas MI size remained constant in placebo and exhibited a trend towards reduction in low dose, high-dose MSC therapy reduced infarct size from 18.2 +/- 0.9 to 14.4 +/- 1.0% (P = 0.02) of LV mass. In addition, both low and high-dose treatments increased regional contractility and MBF in both infarct and border zones. Ectopic tissue formation was not observed with MSCs. CONCLUSION: Together these data demonstrate that autologous MSCs can be safely delivered in an adult heart failure model, producing substantial structural and functional reverse remodelling. These findings demonstrate the safety and efficacy of autologous MSC therapy and support clinical trials of MSC therapy in patients with chronic ischaemic cardiomyopathy.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Animals , Balloon Occlusion , Biomarkers/metabolism , Cytokines/metabolism , Double-Blind Method , Female , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/therapy , Magnetic Resonance Angiography , Myocardial Contraction , Myocardial Infarction/pathology , Myocardial Reperfusion , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/therapy , Myocarditis/blood , Random Allocation , Swine , Transplantation, Autologous , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/therapy , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...