Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Tissue Eng Part A ; 16(8): 2627-39, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20367286

ABSTRACT

Mesenchymal stem cells/marrow stromal cells (MSC) are adult multipotent cells that can augment tissue repair. We previously demonstrated that culturing MSC in hypoxic conditions causes upregulation of the hepatocyte growth factor (HGF) receptor c-Met, allowing them to respond more robustly to HGF. MSC preconditioned in hypoxic environments contributed to restoration of blood flow after an ischemic injury more rapidly than MSC cultured in normoxic conditions. We now investigated the specific role of HGF/c-Met signaling in MSC function. An shRNA-mediated knockdown (KD) of c-Met in MSC did not alter their phenotypic profile, proliferation, or viability in vitro. However, we determined that while HGF/c-Met signaling does not play a role in the adipogenic differentiation of the cells, the disruption of this signaling pathway inhibited the ability of MSC to differentiate into the osteogenic and chondrogenic lineages. We next assessed the impact of c-Met KD on human MSC function in a xenogeneic hindlimb ischemia injury model. A 70% KD of c-Met in MSC resulted in a significant decrease in their capacity to regenerate blood flow to the ischemic limb, as compared to the MSC transduced with control shRNA. MSC with only a 60% KD of c-Met exhibited an intermediate capacity to restore blood flow, suggesting that MSC function is sensitive to the dosage of c-Met signaling. The current study highlights the significance of HGF/c-Met signaling in the capacity of MSC to restore blood flow after an ischemic injury and in their ability to differentiate into the osteogenic and chondrogenic lineages.


Subject(s)
Hindlimb/blood supply , Ischemia/surgery , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering/genetics , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Feasibility Studies , Genetic Enhancement/methods , Hindlimb/surgery , Humans , Ischemia/metabolism , Mice , Mice, SCID , Proto-Oncogene Proteins c-met/genetics , Regeneration/physiology , Treatment Outcome , Up-Regulation/physiology
2.
J Transl Med ; 8: 24, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20214792

ABSTRACT

UNLABELLED: Human stem cells from adult sources have been shown to contribute to the regeneration of muscle, liver, heart, and vasculature. The mechanisms by which this is accomplished are, however, still not well understood. We tested the engraftment and regenerative potential of human umbilical cord blood-derived ALDH(hi)Lin(-), and ALDH(lo)Lin(-) cells following transplantation to NOD/SCID or NOD/SCID beta2m null mice with experimentally induced acute myocardial infarction. We used combined nanoparticle labeling and whole organ fluorescent imaging to detect human cells in multiple organs 48 hours post transplantation. Engraftment and regenerative effects of cell treatment were assessed four weeks post transplantation. We found that ALDH(hi)Lin(-) stem cells specifically located to the site of injury 48 hours post transplantation and engrafted the infarcted heart at higher frequencies than ALDH(lo)Lin(-) committed progenitor cells four weeks post transplantation. We found no donor derived cardiomyocytes and few endothelial cells of donor origin. Cell treatment was not associated with any detectable functional improvement at the four week endpoint. There was, however, a significant increase in vascular density in the central infarct zone of ALDH(hi)Lin(-) cell-treated mice, as compared to PBS and ALDH(lo)Lin(-) cell-treated mice. CONCLUSIONS: Our data indicate that adult human stem cells do not become a significant part of the regenerating tissue, but rapidly home to and persist only temporarily at the site of hypoxic injury to exert trophic effects on tissue repair thereby enhancing vascular recovery.


Subject(s)
Adult Stem Cells/enzymology , Aldehyde Dehydrogenase/metabolism , Fetal Blood , Myocardial Infarction , Neovascularization, Physiologic/physiology , Animals , Cell Lineage , Cell Separation , Fetal Blood/cytology , Fetal Blood/enzymology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/cytology , Myocardium/metabolism , Regeneration/physiology , Stem Cell Transplantation
3.
Biochem Biophys Res Commun ; 372(1): 230-5, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18482578

ABSTRACT

The aim of this study was to create a gene expression profile to better define the phenotype of human adipose-derived stromal cells (HADSCs) during in vitro chondrogenesis, osteogenesis and adipogenesis. A novel aspect of this work was the analysis of the same subset of genes during HADSC differentiation into all three lineages. Chondrogenic induction resulted in increased mRNA expression of Sox transcription factors, COL2A1,COL10A1, Runx2, and Osterix. This is the first report demonstrating significant upregulation in expression of osteogenesis-related transcription factors Runx2 and Osterix by TGF-beta3 induction of HADSCs during in vitro chondrogenesis. These findings suggest that the commonly-used chondrogenic induction reagents promote differentiation suggestive of hypertrophic chondrocytes and osteoblasts. We conclude that alternative strategies are required to induce efficient articular chondrocyte differentiation in order for HADSCs to be of clinical use in cartilage tissue engineering.


Subject(s)
Adipose Tissue/cytology , Chondrocytes/metabolism , Chondrogenesis/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Profiling , Transcription Factors/genetics , Adipogenesis/genetics , Adipose Tissue/drug effects , Adult , Alkaline Phosphatase/genetics , CCAAT-Enhancer-Binding Protein-alpha/genetics , Collagen Type II/genetics , Collagen Type X/genetics , DNA-Binding Proteins/genetics , Female , Hedgehog Proteins/genetics , High Mobility Group Proteins/genetics , Humans , Male , Nuclear Proteins/genetics , Osteogenesis/genetics , PPAR gamma/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , SOX9 Transcription Factor , SOXD Transcription Factors , Sp7 Transcription Factor , Stromal Cells/cytology , Stromal Cells/drug effects , Transforming Growth Factor beta3/pharmacology , Up-Regulation
4.
Stem Cells ; 26(8): 2173-82, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511601

ABSTRACT

Mesenchymal stem cells (MSC) are adult multipotent cells found in bone marrow, adipose tissue, and other adult tissues. MSC have been shown to improve regeneration of injured tissues in vivo, but the mechanisms remain unclear. Typically, MSC are cultured under ambient, or normoxic, conditions (21% oxygen). However, the physiological niches for MSC in the bone marrow and other sites have much lower oxygen tension. When used as a therapeutic tool to repair tissue injuries, MSC cultured in standard conditions must adapt from 21% oxygen in culture to less than 1% oxygen in the ischemic tissue. We therefore examined the effects of preculturing human bone marrow-derived MSC in hypoxic conditions (1%-3% oxygen) to elucidate the best conditions that enhance their tissue regenerative potential. We demonstrated that MSC cultured in hypoxia activate the Akt signaling pathway while maintaining their viability and cell cycle rates. We also showed that MSC cultured in hypoxia induced expression of cMet, the major receptor for hepatocyte growth factor (HGF), and enhanced cMet signaling. MSC cultured in hypoxic conditions increased their migration rates. Since migration and HGF responsiveness are thought to be key mediators of MSC recruitment and/or activation in vivo, we next examined the tissue regenerative potential of MSC cultured under hypoxic conditions, using a murine hind limb ischemia model. We showed that local expression of HGF is increased in ischemic muscle in this model. Intra-arterial injection of MSC cultured in either normoxic or hypoxic conditions 24 hours after surgical induction of hind limb ischemia enhanced revascularization compared with saline controls. However, restoration of blood flow was observed significantly earlier in mice that had been injected with hypoxic preconditioned MSC. Collectively, these data suggest that preculturing MSC under hypoxic conditions prior to transplantation improves their tissue regenerative potential. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Hypoxia , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Regeneration , Animals , Apoptosis , Cell Movement , Humans , Ischemia , Mice , Muscles/metabolism , Oxygen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction
5.
Cell ; 111(1): 51-62, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12372300

ABSTRACT

The therapeutic value of DNA-damaging antineoplastic agents is dependent upon their ability to induce tumor cell apoptosis while sparing most normal tissues. Here, we show that a component of the apoptotic response to these agents in several different types of tumor cells is the deamidation of two asparagines in the unstructured loop of Bcl-xL, and we demonstrate that deamidation of these asparagines imports susceptibility to apoptosis by disrupting the ability of Bcl-xL to block the proapoptotic activity of BH3 domain-only proteins. Conversely, Bcl-xL deamidation is actively suppressed in fibroblasts, and suppression of deamidation is an essential component of their resistance to DNA damage-induced apoptosis. Our results suggest that the regulation of Bcl-xL deamidation has a critical role in the tumor-specific activity of DNA-damaging antineoplastic agents.


Subject(s)
DNA Damage , Gene Expression Regulation , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Apoptosis , Asparagine/chemistry , Asparagine/metabolism , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Humans , Immunoblotting , Microscopy, Fluorescence , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , Peptides/chemistry , Plasmids/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Time Factors , Transfection , Tumor Cells, Cultured , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...