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











Publication year range
1.
Arterioscler Thromb Vasc Biol ; 37(2): 280-290, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27908889

ABSTRACT

OBJECTIVE: Lineage-negative bone marrow cells (lin- BMCs) are enriched in endothelial progenitor cells and mediate vascular repair. Aging-associated senescence and apoptosis result in reduced number and functionality of lin- BMCs, impairing their prorepair capacity. The molecular mechanisms underlying lin- BMC senescence and apoptosis are poorly understood. MicroRNAs (miRNAs) regulate many important biological processes. The identification of miRNA-mRNA networks that modulate the health and functionality of lin- BMCs is a critical step in understanding the process of vascular repair. The aim of this study was to characterize the role of the miR-146a-Polo-like kinase 2 (Plk2) network in regulating lin- BMC senescence, apoptosis, and their angiogenic capability. APPROACH AND RESULTS: Transcriptome analysis in lin- BMCs isolated from young and aged wild-type and ApoE-/- (apolipoprotein E) mice showed a significant age-associated increase in miR-146a expression. In silico analysis, expression study and Luciferase reporter assay established Plk2 as a direct target of miR-146a. miR-146a overexpression in young lin- BMCs inhibited Plk2 expression, resulting in increased senescence and apoptosis, via p16Ink4a/p19Arf and p53, respectively, as well as impaired angiogenic capacity in vitro and in vivo. Conversely, suppression of miR-146a in aged lin- BMCs increased Plk2 expression and rejuvenated lin- BMCs, resulting in decreased senescence and apoptosis, leading to improved angiogenesis. CONCLUSIONS: (1) miR-146a regulates lin- BMC senescence and apoptosis by suppressing Plk2 expression that, in turn, activates p16Ink4a/p19Arf and p53 and (2) modulation of miR-146a or its target Plk2 may represent a potential therapeutic intervention to improve lin- BMC-mediated angiogenesis and vascular repair.


Subject(s)
Apoptosis , Bone Marrow Cells/enzymology , Cell Lineage , Cellular Senescence , Endothelial Progenitor Cells/enzymology , MicroRNAs/metabolism , Protein Serine-Threonine Kinases/metabolism , 3' Untranslated Regions , Age Factors , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Binding Sites , Bone Marrow Cells/pathology , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation , Endothelial Progenitor Cells/pathology , Genotype , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Neovascularization, Physiologic , Phenotype , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Signal Transduction , Transcriptome , Transfection , Tumor Suppressor Protein p53/metabolism
2.
Biochem Biophys Res Commun ; 430(2): 827-32, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23201405

ABSTRACT

HDAC inhibitors are under clinical development for the treatment of hypertrophic cardiomyopathy and heart failure although the mechanisms of protection are incompletely understood. Micro-RNA 126, an endothelium-specific miR has been assigned essential developmental roles in the heart by activating survival kinases ERK1/2 and Akt and increasing pro-angiogenic signaling. Here we provide the first evidence that hypoxia and HDAC inhibitors selectively and synergistically stimulate expression of miR-126 in cardiac myocytes. MiR-126 expression was increased 1.7-fold (p<0.05) after 1h of hypoxic exposure and this was further enhanced to 3.0-fold (p<0.01) by simultaneously blocking HDAC with the pan-HDAC inhibitor Tricostatin A (TSA). TSA alone did not increase miR-126. In parallel, hypoxia and TSA synergistically increased p-ERK and p-Akt without effecting VEGF-A level. Knockdown of miR-126 with si-RNA eliminated inductions of p-ERK and p-Akt by hypoxia, whereas miR-126 overexpression mimicked hypoxia and amplified p-ERK and p-Akt in parallel with miR-126. The results suggest that miR-126 is a hypoxia-inducible target of HAT/HDAC and its activation in cardiac myocytes may contribute to cardioprotection by activating cell survival and pro-angiogenic pathways selectively during ischemia.


Subject(s)
Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , MicroRNAs/biosynthesis , Myocytes, Cardiac/enzymology , Animals , Cell Hypoxia , Cells, Cultured , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , MicroRNAs/genetics , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats
3.
Circ Res ; 112(1): 152-64, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23072816

ABSTRACT

RATIONALE: Endothelial progenitor cells (EPCs) contribute to the regeneration of endothelium. Aging-associated senescence results in reduced number and function of EPCs, potentially contributing to increased cardiac risk, reduced angiogenic capacity, and impaired cardiac repair effectiveness. The mechanisms underlying EPC senescence are unknown. Increasing evidence supports the role of microRNAs in regulating cellular senescence. OBJECTIVE: We aimed to determine whether microRNAs regulated EPC senescence and, if so, what the underlying mechanisms are. METHODS AND RESULTS: To map the microRNA/gene expression signatures of EPC senescence, we performed microRNA profiling and microarray analysis in lineage-negative bone marrow cells from young and aged wild-type and apolipoprotein E-deficient mice. We identified 2 microRNAs, microRNA-10A* (miR-10A*), and miR-21, and their common target gene Hmga2 as critical regulators for EPC senescence. Overexpression of miR-10A* and miR-21 in young EPCs suppressed Hmga2 expression, caused EPC senescence, as evidenced by senescence-associated ß-galactosidase upregulation, decreased self-renewal potential, increased p16(Ink4a)/p19(Arf) expression, and resulted in impaired EPC angiogenesis in vitro and in vivo, resembling EPCs derived from aged mice. In contrast, suppression of miR-10A* and miR-21 in aged EPCs increased Hmga2 expression, rejuvenated EPCs, resulting in decreased senescence-associated ß-galactosidase expression, increased self-renewal potential, decreased p16(Ink4a)/p19(Arf) expression, and improved EPC angiogenesis in vitro and in vivo. Importantly, these phenotypic changes were rescued by miRNA-resistant Hmga2 cDNA overexpression. CONCLUSIONS: miR-10A* and miR-21 regulate EPC senescence via suppressing Hmga2 expression and modulation of microRNAs may represent a potential therapeutic intervention in improving EPC-mediated angiogenesis and vascular repair.


Subject(s)
Cellular Senescence , Endothelial Cells/metabolism , HMGB3 Protein/metabolism , MicroRNAs/metabolism , Stem Cells/metabolism , Aging/genetics , Aging/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cell Proliferation , Cells, Cultured , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Down-Regulation , Gene Expression Profiling/methods , Genotype , HMGB3 Protein/genetics , Hindlimb , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Oligonucleotide Array Sequence Analysis , Phenotype , Transfection , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
4.
Atherosclerosis ; 212(1): 63-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20451204

ABSTRACT

AIMS: Vein graft endothelial damage is a key step in the development of neointimal hyperplasia, leading to vein graft failure. We sought to determine whether exogenous endothelial progenitor cells could promote vein graft re-endothelialization, and thereby ameliorate neointimal hyperplasia. METHODS AND RESULTS: Carotid artery interposition grafting was performed with syngeneic inferior vena cavae in mice with severe combined immunodeficiency (SCID). Lineage-negative human umbilical cord blood (hUCB) cells (or medium alone) were injected into vein-grafted mice intra-operatively and 2 weeks post-operatively. In vein grafts from hUCB cell-injected mice, we found human HLA-expressing endothelial cells, as well as increased levels of VEGF and FGF-2. Furthermore, hUCB cells secreted VEGF and FGF-2 in vitro. The markedly enhanced endothelial regeneration, likely resulting from both direct engraftment and paracrine actions of hUCB cells, inhibited inflammatory response, diminished intimal cell proliferation, and reduced neointimal hyperplasia in the vein grafts. CONCLUSIONS: hUCB cells may accelerate vein graft re-endothelialization via both direct differentiation into endothelial cells and release of paracrine factors to enhance endothelial regeneration and reduce inflammation. These data highlight a potential therapeutic role for cellular therapy in vessel injury.


Subject(s)
Carotid Arteries/surgery , Cord Blood Stem Cell Transplantation , Endothelial Cells/transplantation , Graft Survival , Vena Cava, Inferior/transplantation , Animals , Carotid Arteries/pathology , Cell Differentiation , Cell Proliferation , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblast Growth Factor 2/metabolism , HLA Antigens/metabolism , Humans , Hyperplasia , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Mice, SCID , Paracrine Communication , Regeneration , Time Factors , Tunica Intima/pathology , Vascular Endothelial Growth Factor A/metabolism , Vena Cava, Inferior/immunology , Vena Cava, Inferior/metabolism , Vena Cava, Inferior/pathology
5.
Proc Natl Acad Sci U S A ; 106(21): 8683-8, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19429708

ABSTRACT

Kaposi's sarcoma (KS) is the major AIDS-associated malignancy. It is characterized by the proliferation of spindle cells, inflammatory infiltrate, and aberrant angiogenesis caused by Kaposi's sarcoma herpesvirus (KSHV) infection. Small GTPase Rac1, an inflammatory signaling mediator triggering reactive oxygen species (ROS) production by NADPH-oxidases, is implicated in carcinogenesis and tumor angiogenesis. Here, we show that expression of a constitutively active Rac1 (RacCA) driven by the alpha-smooth muscle actin promoter in transgenic mice is sufficient to cause KS-like tumors through mechanisms involving ROS-driven proliferation, up-regulation of AKT signaling, and hypoxia-inducible factor 1-alpha-related angiogenesis. RacCA-induced tumors expressed KS phenotypic markers; displayed remarkable transcriptome overlap with KS lesions; and were, like KS, associated with male gender. The ROS scavenging agent N-acetyl-cysteine inhibited angiogenesis and completely abrogated transgenic RacCA tumor formation, indicating a causal role of ROS in tumorigenesis. Consistent with a pathogenic role in KS, immunohistochemical analysis revealed that Rac1 is overexpressed in KSHV(+) spindle cells of AIDS-KS biopsies. Our results demonstrate the direct oncogenicity of Rac1 and ROS and their contribution to a KS-like malignant phenotype, further underscoring the carcinogenic potential of oxidative stress in the context of chronic infection and inflammation. They define the RacCA transgenic mouse as a model suitable for studying the role of oxidative stress in the pathogenesis and therapy of KS, with relevance to other inflammation-related malignancies. Our findings suggest host and viral genes triggering Rac1 or ROS production as key determinants of KS onset and potential KS chemopreventive or therapeutic targets.


Subject(s)
Antioxidants/metabolism , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , rac1 GTP-Binding Protein/metabolism , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/enzymology , Acquired Immunodeficiency Syndrome/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/etiology , Transcription, Genetic/genetics , Tumor Cells, Cultured , rac1 GTP-Binding Protein/genetics
6.
Circulation ; 118(21): 2156-65, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-18981303

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) contribute to vascular regeneration/repair and thus may protect against scleroderma vasculopathy. We aimed to determine whether circulating EPCs were reduced in scleroderma, whether scleroderma sera could induce EPC apoptosis, and, if so, what the underlying apoptotic signaling pathway was. METHODS AND RESULTS: Circulating EPC levels were quantified in 54 patients with scleroderma and 18 healthy control subjects by colony-forming unit assay and flow cytometry, which revealed markedly decreased EPC levels in scleroderma patients relative to healthy subjects. Substantial apoptosis was detected in EPCs after culturing in the presence of scleroderma sera compared with normal sera. Intriguingly, depletion of the IgG fraction from scleroderma sera completely abolished the apoptotic effects. Furthermore, scleroderma sera inhibited the activation/phosphorylation of Akt, which in turn suppressed the phosphorylation and degradation of forkhead transcription factor FKHRL1 (FOXO3a), resulting in the upregulation of apoptotic protein Bim. siRNA-mediated FOXO3a and Bim knockdown substantially reduced scleroderma serum-induced EPC apoptosis. Importantly, Bim expression and baseline apoptosis were increased in EPCs freshly isolated from scleroderma patients relative to that obtained from healthy subjects. CONCLUSIONS: Scleroderma serum-induced EPC apoptosis is mediated chiefly by the Akt-FOXO3a-Bim pathway, which may account, at least in part, for the decreased circulating EPC levels in scleroderma patients.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis , Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Membrane Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/biosynthesis , Scleroderma, Systemic/metabolism , Stem Cells/metabolism , Transcription, Genetic , Up-Regulation , Adult , Aged , Bcl-2-Like Protein 11 , Cells, Cultured , Colony-Forming Units Assay/methods , Endothelial Cells/pathology , Female , Forkhead Box Protein O3 , Humans , Immunoglobulin G/metabolism , Male , Middle Aged , Regeneration , Scleroderma, Systemic/pathology , Serum/metabolism , Stem Cells/pathology
7.
J Am Coll Cardiol ; 50(23): 2243-8, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18061073

ABSTRACT

OBJECTIVES: Our objective was to develop and assess a novel endogenous progenitor cell (EPC) assay based on aldehyde dehydrogenase (ALDH) activity, and to define the relationship of ALDH-bright (ALDH(br)) cells with previously defined EPCs, patient age, and extent of coronary artery disease. BACKGROUND: Accurate assessment of circulating EPCs is of significant interest, yet current assays have limitations. Progenitor cells display high levels of ALDH activity. An assay based on ALDH activity may offer a simple means for enumerating EPCs. METHODS: We simultaneously determined the numbers of EPCs based on ALDH activity and cell surface expression of CD133, CD34, and vascular endothelial growth factor receptor-2 in 110 patients undergoing cardiac catheterization. We assessed the reproducibility of these estimates, correlation among EPC assays, and the association of ALDH(br) numbers with age and disease severity. RESULTS: Aldehyde dehydrogenase-bright cells were easily identified in nonmobilized peripheral blood with median and mean frequencies of 0.041% and 0.074%, respectively. Aldehyde dehydrogenase-bright cells expressed CD34 or CD133 cell surface markers (57.0% and 27.1%, respectively), correlated closely with CD133+CD34+ cells (r = 0.72; p < 0.001), and differentiated into endothelial cells with greater efficiency than CD133+CD34+ cells. Aldehyde dehydrogenase-bright cell numbers were inversely associated with patient age and coronary disease severity. CONCLUSIONS: Aldehyde dehydrogenase activity represents a novel simplified method for quantifying EPCs. The correlation of ALDH(br) cells with clinical factors and outcomes warrants further study.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Cell Count/methods , Coronary Artery Disease/enzymology , Leukocytes, Mononuclear/enzymology , Stem Cells/enzymology , AC133 Antigen , Age Factors , Aged , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cardiac Catheterization , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Female , Glycoproteins/metabolism , Humans , Male , Middle Aged , Peptides/metabolism , Reproducibility of Results , Severity of Illness Index , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Arterioscler Thromb Vasc Biol ; 27(1): 113-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17095715

ABSTRACT

OBJECTIVE: We have demonstrated that bone marrow cells from young and wild-type (WT), but not old apoE-/-, mice are capable of preventing atherosclerosis. This study was performed to elucidate the numerical and functional changes underlying the efficacy difference between young and old bone marrow. METHODS AND RESULTS: CD34+/VEGFR2+ conventional endothelial progenitor cells and lin-/cKit+/Sca-1+ hematopoietic stem cells did not differ numerically or functionally between young and old apoE-/- bone marrow. Fluorescence-activated cell sorter analysis, however, showed that a group of cells (simple little cells or SLCs), characteristically located in the lower left quadrant of forward scatter/side scatter flow cytometric plot, were markedly decreased in old WT and apoE-/- marrow, but abundantly present in young WT and apoE-/- bone marrow. The SLC fraction was mainly composed of lin-/cKit-/Sca-1- cells. In vitro differentiation assay demonstrated substantially more efficient endothelial differentiation of lin-/cKit-/Sca-1- SLCs than other bone marrow fractions at a single cell level and en masse. Furthermore, old lin-/cKit-/Sca-1- SLCs had a trend of decreased endothelial differentiation capability. CONCLUSIONS: Lin-/cKit-/Sca-1- SLCs may represent a previously unrecognized cell population, enriched for endothelial progenitors. The identification of these cells may help improve the efficacy of cell therapy.


Subject(s)
Atherosclerosis/physiopathology , Bone Marrow/pathology , Cellular Senescence/physiology , Endothelium/pathology , Hematopoietic Stem Cells/pathology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/pathology , Bone Marrow/metabolism , Bone Marrow/physiopathology , Cell Count , Cell Differentiation , Cells, Cultured , Endothelium/metabolism , Endothelium/physiopathology , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism
9.
PLoS Med ; 2(12): e354, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16250671

ABSTRACT

BACKGROUND: Systemic sclerosis (SSc) is characterized by excessive fibrosis and obliterative vascular lesions. Abnormal TGFbeta activation is implicated in the pathogenesis of SSc. Aberrant TGFbeta/Smad signaling can be controlled by stabilization of microtubules with paclitaxel. METHODS AND FINDINGS: SSc and healthy human skin biopsies were incubated in the presence or absence of paclitaxel followed by transplantation into severe combined immunodeficient mice. TGFbeta signaling, fibrosis, and neovessel formation were evaluated by quantitative RT-PCR and immunohistochemical staining. Paclitaxel markedly suppressed Smad2 and Smad3 phosphorylation and collagen deposition in SSc grafts. As a result, the autonomous maintenance/reconstitution of the SSc phenotype was prevented. Remarkably, SSc grafts showed a 2-fold increase in neovessel formation relative to normal grafts, regardless of paclitaxel treatment. Angiogenesis in SSc grafts was associated with a substantial increase in mouse PECAM-1 expression, indicating the mouse origin of the neovascular cells. CONCLUSION: Low-dose paclitaxel can significantly suppress TGFbeta/Smad activity and lessen fibrosis in SCID mice. Transplantation of SSc skin into SCID mice elicits a strong angiogenesis-an effect not affected by paclitaxel. Although prolonged chemotherapy with paclitaxel at higher doses is associated with pro-fibrotic and anti-angiogenic changes, the findings described here indicate that low-dose paclitaxel may have therapeutic benefits for SSc via modulating TGFbeta signaling.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/pharmacology , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/physiopathology , Signal Transduction/drug effects , Transforming Growth Factor beta/physiology , Adult , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Biopsy , Dose-Response Relationship, Drug , Female , Fibrosis , Humans , Male , Mice , Mice, SCID , Middle Aged , Neovascularization, Pathologic , Paclitaxel/administration & dosage , Phenotype , Smad Proteins/drug effects , Smad Proteins/physiology , Transforming Growth Factor beta/drug effects , Transplantation, Heterologous
10.
Nat Med ; 11(9): 952-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16142243

ABSTRACT

Nitric oxide (NO) production by endothelial cell nitric oxide synthase (eNOS) in sinusoidal endothelial cells is reduced in the injured liver and leads to intrahepatic portal hypertension. We sought to understand the mechanism underlying defective eNOS function. Phosphorylation of the serine-threonine kinase Akt, which activates eNOS, was substantially reduced in sinusoidal endothelial cells from injured livers. Overexpression of Akt in vivo restored phosphorylation of Akt and production of NO and reduced portal pressure in portal hypertensive rats. We found that Akt physically interacts with G-protein-coupled receptor kinase-2 (GRK2), and that this interaction inhibits Akt activity. Furthermore, GRK2 expression increased in sinusoidal endothelial cells from portal hypertensive rats and knockdown of GRK2 restored Akt phosphorylation and NO production, and normalized portal pressure. Finally, after liver injury, GRK2-deficient mice developed less severe portal hypertension than control mice. Thus, an important mechanism underlying impaired activity of eNOS in injured sinusoidal endothelial cells is defective phosphorylation of Akt caused by overexpression of GRK2 after injury.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Endothelial Cells/enzymology , Hypertension, Portal/metabolism , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2 , Isoenzymes , Male , Nitric Oxide , Nitric Oxide Synthase Type III , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , beta-Adrenergic Receptor Kinases
SELECTION OF CITATIONS
SEARCH DETAIL