Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cell Microbiol ; 17(10): 1447-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25857345

ABSTRACT

The contribution of myeloid cells to tumour microenvironments is a decisive factor in cancer progression. Tumour-associated macrophages (TAMs) mediate tumour invasion and angiogenesis through matrix remodelling, immune modulation and release of pro-angiogenic cytokines. Nothing is known about how pathogenic bacteria affect myeloid cells in these processes. Here we show that Bartonella henselae, a bacterial pathogen causing vasculoproliferative diseases (bacillary angiomatosis), reprogrammes human myeloid angiogenic cells (MACs), a pro-angiogenic subset of circulating progenitor cells, towards a TAM-like phenotype with increased pro-angiogenic capacity. B. henselae infection resulted in inhibition of cell death, activation of angiogenic cellular programmes and induction of M2 macrophage polarization. MACs infected with B. henselae incorporated into endothelial sprouts and increased angiogenic growth. Infected MACs developed a vascular mimicry phenotype in vitro, and expression of B. henselae adhesin A was essential in inducing these angiogenic effects. Secretome analysis revealed that increased pro-angiogenic activities were associated with the creation of a tumour-like microenvironment dominated by angiogenic inflammatory cytokines and matrix remodelling compounds. Our results demonstrate that manipulation of myeloid cells by pathogenic bacteria can contribute to microenvironmental regulation of pathological tissue growth and suggest parallels underlying both bacterial infections and cancer.


Subject(s)
Bartonella henselae/physiology , Host-Pathogen Interactions , Myeloid Progenitor Cells/physiology , Neovascularization, Pathologic , Cell Differentiation , Endothelial Cells/microbiology , Endothelial Cells/physiology , Humans , Macrophages/microbiology , Macrophages/physiology
2.
Arterioscler Thromb Vasc Biol ; 33(3): 533-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23288173

ABSTRACT

OBJECTIVE: Histone deacetylases (HDACs) modulate gene expression by deacetylation of histone and nonhistone proteins. Several HDACs control angiogenesis, but the role of HDAC9 is unclear. METHODS AND RESULTS: Here, we analyzed the function of HDAC9 in angiogenesis and its involvement in regulating microRNAs. In vitro, silencing of HDAC9 reduces endothelial cell tube formation and sprouting. Furthermore, HDAC9 silencing decreases vessel formation in a spheroid-based Matrigel plug assay in mice and disturbs vascular patterning in zebrafish embryos. Genetic deletion of HDAC9 reduces retinal vessel outgrowth and impairs blood flow recovery after hindlimb ischemia. Consistently, overexpression of HDAC9 increases endothelial cell sprouting, whereas mutant constructs lacking the catalytic domain, the nuclear localization sequence, or sumoylation site show no effect. To determine the mechanism underlying the proangiogenic effect of HDAC9, we measured the expression of the microRNA (miR)-17-92 cluster, which is known for its antiangiogenic activity. We demonstrate that silencing of HDAC9 in endothelial cells increases the expression of miR-17-92. Inhibition of miR-17-20a rescues the sprouting defects induced by HDAC9 silencing in vitro and blocking miR-17 expression partially reverses the disturbed vascular patterning of HDAC9 knockdown in zebrafish embryos. CONCLUSIONS: We found that HDAC9 promotes angiogenesis and transcriptionally represses the miR-17-92 cluster.


Subject(s)
Histone Deacetylases/metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Ischemia/enzymology , MicroRNAs/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Repressor Proteins/metabolism , Retinal Neovascularization/enzymology , Zebrafish Proteins/metabolism , Animals , Disease Models, Animal , Gene Knockdown Techniques , HEK293 Cells , Hindlimb , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Humans , Ischemia/genetics , Ischemia/physiopathology , Mice , Mice, Knockout , MicroRNAs/genetics , Mutation , Neovascularization, Physiologic/genetics , RNA Interference , RNA, Long Noncoding , Regional Blood Flow , Repressor Proteins/deficiency , Repressor Proteins/genetics , Retinal Neovascularization/genetics , Retinal Neovascularization/physiopathology , Transfection , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Nat Cell Biol ; 14(3): 249-56, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327366

ABSTRACT

The shear-responsive transcription factor Krüppel-like factor 2 (KLF2) is a critical regulator of endothelial gene expression patterns induced by atheroprotective flow. As microRNAs (miRNAs) post-transcriptionally control gene expression in many pathogenic and physiological processes, we investigated the regulation of miRNAs by KLF2 in endothelial cells. KLF2 binds to the promoter and induces a significant upregulation of the miR-143/145 cluster. Interestingly, miR-143/145 has been shown to control smooth muscle cell (SMC) phenotypes; therefore, we investigated the possibility of transport of these miRNAs between endothelial cells and SMCs. Indeed, extracellular vesicles secreted by KLF2-transduced or shear-stress-stimulated HUVECs are enriched in miR-143/145 and control target gene expression in co-cultured SMCs. Extracellular vesicles derived from KLF2-expressing endothelial cells also reduced atherosclerotic lesion formation in the aorta of ApoE(-/-) mice. Combined, our results show that atheroprotective stimuli induce communication between endothelial cells and SMCs through an miRNA- and extracellular-vesicle-mediated mechanism and that this may comprise a promising strategy to combat atherosclerosis.


Subject(s)
Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/transplantation , Exosomes/metabolism , Exosomes/transplantation , Exosomes/ultrastructure , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Mice , Mice, Knockout , MicroRNAs/metabolism , Microscopy, Confocal , Microscopy, Electron , Myocytes, Smooth Muscle/cytology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Stress, Mechanical , Transfection
4.
Blood ; 119(6): 1607-16, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22184411

ABSTRACT

MicroRNAs (miRs) are small RNAs that regulate gene expression at the posttranscriptional level. miR-27 is expressed in endothelial cells, but the specific functions of miR-27b and its family member miR-27a are largely unknown. Here we demonstrate that overexpression of miR-27a and miR-27b significantly increased endothelial cell sprouting. Inhibition of both miR-27a and miR-27b impaired endothelial cell sprout formation and induced endothelial cell repulsion in vitro. In vivo, inhibition of miR-27a/b decreased the number of perfused vessels in Matrigel plugs and impaired embryonic vessel formation in zebrafish. Mechanistically, miR-27 regulated the expression of the angiogenesis inhibitor semaphorin 6A (SEMA6A) in vitro and in vivo and targeted the 3'-untranslated region of SEMA6A. Silencing of SEMA6A partially reversed the inhibition of endothelial cell sprouting and abrogated the repulsion of endothelial cells mediated by miR-27a/b inhibition, indicating that SEMA6A is a functionally relevant miR-27 downstream target regulating endothelial cell repulsion. In summary, we show that miR-27a/b promotes angiogenesis by targeting the angiogenesis inhibitor SEMA6A, which controls repulsion of neighboring endothelial cells.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/genetics , Neovascularization, Physiologic/genetics , Semaphorins/genetics , 3' Untranslated Regions/genetics , Animals , Blood Vessels/embryology , Blood Vessels/metabolism , Blotting, Western , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Endothelial Cells/physiology , Gene Expression , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Neovascularization, Physiologic/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Semaphorins/metabolism , Transfection , Zebrafish/embryology , Zebrafish/genetics
5.
Circ Res ; 109(11): 1219-29, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21980126

ABSTRACT

RATIONALE: Proangiogenic hematopoietic and endothelial progenitor cells (EPCs) contribute to postnatal neovascularization, but the mechanisms regulating differentiation to the endothelial lineage are unclear. OBJECTIVE: To elucidate the epigenetic control of endothelial gene expression in proangiogenic cells and EPCs. METHODS AND RESULTS: Here we demonstrate that the endothelial nitric oxide synthase (eNOS) promoter is epigenetically silenced in proangiogenic cells (early EPCs), CD34(+) cells, and mesoangioblasts by DNA methylation and prominent repressive histone H3K27me3 marks. In order to reverse epigenetic silencing to facilitate endothelial commitment, we used 3-deazaneplanocin A, which inhibits the histone methyltransferase enhancer of zest homolog 2 and, thereby, reduces H3K27me3. 3-Deazaneplanocin A was not sufficient to increase eNOS expression, but the combination of 3-deazaneplanocin A and the histone deacetylase inhibitor Trichostatin A augmented eNOS expression, indicating that the concomitant inhibition of silencing histone modification and enhancement of activating histone modification facilitates eNOS expression. In ischemic tissue, hypoxia plays a role in recruiting progenitor cells. Therefore, we examined the effect of hypoxia on epigenetic modifications. Hypoxia modulated the balance of repressive to active histone marks and increased eNOS mRNA expression. The reduction of repressive H3K27me3 was associated with an increase of the histone demethylase Jmjd3. Silencing of Jmjd3 induced apoptosis and senescence in proangiogenic cells and inhibited hypoxia-mediated up-regulation of eNOS expression in mesoangioblasts. CONCLUSIONS: These findings provide evidence that histone modifications epigenetically control the eNOS promoter in proangiogenic cells.


Subject(s)
DNA Methylation/physiology , Endothelial Cells/cytology , Hematopoietic Stem Cells/physiology , Neovascularization, Physiologic/genetics , Nitric Oxide Synthase Type III/genetics , Acetylation/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Apoptosis/drug effects , Cell Hypoxia/genetics , Cell Lineage , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cellular Senescence/drug effects , DNA Methylation/drug effects , Enzyme Induction/drug effects , Hematopoietic Stem Cells/cytology , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Jumonji Domain-Containing Histone Demethylases/physiology , Nitric Oxide Synthase Type III/biosynthesis , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
6.
EMBO J ; 30(20): 4142-56, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21847094

ABSTRACT

Histone deacetylases (HDACs) deacetylate histones and non-histone proteins, thereby affecting protein activity and gene expression. The regulation and function of the cytoplasmic class IIb HDAC6 in endothelial cells (ECs) is largely unexplored. Here, we demonstrate that HDAC6 is upregulated by hypoxia and is essential for angiogenesis. Silencing of HDAC6 in ECs decreases sprouting and migration in vitro and formation of functional vascular networks in matrigel plugs in vivo. HDAC6 regulates zebrafish vessel formation, and HDAC6-deficient mice showed a reduced formation of perfused vessels in matrigel plugs. Consistently, overexpression of wild-type HDAC6 increases sprouting from spheroids. HDAC6 function requires the catalytic activity but is independent of ubiquitin binding and deacetylation of α-tubulin. Instead, we found that HDAC6 interacts with and deacetylates the actin-remodelling protein cortactin in ECs, which is essential for zebrafish vessel formation and which mediates the angiogenic effect of HDAC6. In summary, we show that HDAC6 is necessary for angiogenesis in vivo and in vitro, involving the interaction and deacetylation of cortactin that regulates EC migration and sprouting.


Subject(s)
Cell Movement , Cortactin/metabolism , Histone Deacetylases/metabolism , Neovascularization, Physiologic , Tubulin/metabolism , Zebrafish Proteins/metabolism , Acetylation , Animals , Carcinoma, Lewis Lung/enzymology , Cells, Cultured , Endothelial Cells/metabolism , Female , Histone Deacetylase 6 , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, Knockout
7.
Clin Res Cardiol ; 100(10): 925-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21633921

ABSTRACT

BACKGROUND: Limited data is available for investigating the long-term safety and effects of intracoronary progenitor cell therapy in patients with acute myocardial infarction (AMI). OBJECTIVE: To assess the clinical course, NT-proBNP and MRI data as objective markers of cardiac function of the TOPCARE-AMI patients at 5-year follow-up. DESIGN: The TOPCARE-AMI trial was the first randomized study investigating the effects of intracoronary infusion of circulating (CPC) or bone marrow-derived progenitor cells (BMC) in 59 patients with successfully reperfused AMI. RESULTS: Five-year follow-up data were completed in 55 patients, 3 patients were lost to follow-up. None of the patients showed any signs of intramyocardial calcification or tumors at 5 years. One patient died during the initial hospitalization, no patient was rehospitalized for heart failure and 16 patients underwent target vessel revascularization (TVR). Only two TVRs occurred later than 1 year after cell administration making it very unlikely that the infused cells accelerate atherosclerotic disease progression. Serum levels of NT-proBNP remained significantly reduced at the 5-year follow-up indicating the absence of heart failure. MRI subgroup analysis in 31 patients documented a persistent improvement of LV ejection fraction (from 46 ± 10% at baseline to 57 ± 10% at 5 years, p < 0.001)). Simultaneously, there was a reduction (p < 0.001) in functional infarct size measured as late enhancement volume normalized to LV mass. However, whereas LV end-systolic volume remained stable, LV end-diastolic volume increased significantly. CONCLUSIONS: The 5-year follow-up of the TOPCARE-AMI trial provides reassurance with respect to the long-term safety of intracoronary cell therapy and suggests favorable effects on LV function.


Subject(s)
Angioplasty, Balloon, Coronary , Myocardial Infarction/therapy , Myocardium/pathology , Regeneration , Stem Cell Transplantation , Adult , Angioplasty, Balloon, Coronary/adverse effects , Angioplasty, Balloon, Coronary/instrumentation , Angioplasty, Balloon, Coronary/mortality , Biomarkers/blood , Combined Modality Therapy , Disease-Free Survival , Germany , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Pilot Projects , Platelet Aggregation Inhibitors/therapeutic use , Recovery of Function , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/mortality , Stents , Stroke Volume , Time Factors , Treatment Outcome , Ventricular Function, Left
8.
J Mol Cell Cardiol ; 50(2): 333-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21147123

ABSTRACT

Early pro-angiogenic cells (EPCs) have been shown to be involved in neovascularization, angiogenesis and re-endothelialization and cathepsin L inhibition blunted their pro-angiogenic effect. In the present study, we have analysed and mapped the proteome and secretome of human EPCs, utilizing a combination of difference in-gel electrophoresis (DIGE) and shotgun proteomics. A population of 206 protein spots were analysed, with 171 being identified in the cellular proteome of EPCs. 82 proteins were identified in their conditioned medium, including the alternative macrophage markers C-C motif chemokine 18 (CCL18) and the hemoglobin scavenger receptor CD163 as well as platelet factor 4 (CXCL4) and platelet basic protein (CXCL7) with "platelet alpha granule" being returned as the top category according to the Gene Ontology Annotation. Apart from cathepsin L, the cathepsin L inhibitor also attenuated the release of a wide range of other cathepsins and lysosomal proteins such as legumain, but stimulated the secretion of members of the S100 protein family. The data presented here are the most comprehensive characterization of protein expression and secretion in human EPCs to date and highlight the potential importance of cysteine proteases in the processing of platelet factors for their pro-angiogenic potential. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".


Subject(s)
Endothelial Cells/metabolism , Proteomics , Blood Platelets/cytology , Cathepsin L/antagonists & inhibitors , Cells, Cultured , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Myoblasts, Smooth Muscle/drug effects , Myoblasts, Smooth Muscle/metabolism
9.
Eur Heart J ; 32(3): 371-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20494899

ABSTRACT

AIMS: Coronary artery disease (CAD) patients have less circulating proangiogenic cells (PACs), formerly known as endothelial progenitor cells, which exhibit impaired neovascularization properties. Inverse correlations were also found between PAC function and risk factors like age. Krüppel-like factor 2 (KLF2) is expressed by mature endothelial cells (ECs), is induced by both shear stress and statins, and provokes endothelial functional differentiation. The aim of this study is to identify whether KLF2 can reverse negative effects of ageing on PAC function. METHODS AND RESULTS: We describe that progenitor cells in the bone marrow and PACs also express KLF2 at a comparable level to mature ECs and that senescence decreases KLF2 levels. To study the effects of ageing on KLF2 levels, we compared progenitor cells of 4 weeks and 16- to 18-month-old C57BL/6 mice. In addition to the three-fold reduction of circulating Sca1(+)/c-Kit(+)/Lin(-) progenitor cells and the 15% reduction of Sca1(+)/Flk1(+) endothelial-committed progenitor cells, the spleen-derived PACs and bone marrow-derived progenitor cells isolated from aged mice showed a lower level of KLF2 when compared with young mice. Lentiviral overexpression of KLF2 increased human PAC numbers and endothelial nitric oxide synthase expression by 60% during in vitro culture. Endothelial lineage-specific KLF2 overexpression in aged bone marrow-derived mononuclear cells strongly augments neovascularization in vivo in a murine hind-limb ischaemia model. CONCLUSION: These results imply that KLF2 is an attractive novel target to rejuvenate PACs before autologous administration to CAD patients.


Subject(s)
Cellular Senescence/physiology , Coronary Artery Disease/physiopathology , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Kruppel-Like Transcription Factors/metabolism , Stem Cells/cytology , Animals , Collateral Circulation/physiology , Hindlimb/blood supply , Ischemia/physiopathology , Leukocytes, Mononuclear/physiology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology
10.
Cancer Res ; 70(13): 5270-80, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530679

ABSTRACT

TIE2-expressing monocytes/macrophages (TEM) are a highly proangiogenic subset of myeloid cells in tumors. Here, we show that circulating human TEMs are already preprogrammed in the circulation to be more angiogenic and express higher levels of such proangiogenic genes as matrix metalloproteinase-9 (MMP-9), VEGFA, COX-2, and WNT5A than TIE2(-) monocytes. Additionally, angiopoietin-2 (ANG-2) markedly enhanced the proangiogenic activity of TEMs and increased their expression of two proangiogenic enzymes: thymidine phosphorylase (TP) and cathepsin B (CTSB). Three "alternatively activated" (or M2-like) macrophage markers were also upregulated by ANG-2 in TEMs: interleukin-10, mannose receptor (MRC1), and CCL17. To investigate the effects of ANG-2 on the phenotype and function of TEMs in tumors, we used a double-transgenic (DT) mouse model in which ANG-2 was specifically overexpressed by endothelial cells. Syngeneic tumors grown in these ANG-2 DT mice were more vascularized and contained greater numbers of TEMs than those in wild-type (WT) mice. In both tumor types, expression of MMP-9 and MRC1 was mainly restricted to tumor TEMs rather than TIE2(-) macrophages. Furthermore, tumor TEMs expressed higher levels of MRC1, TP, and CTSB in ANG-2 DT tumors than WT tumors. Taken together, our data show that although circulating TEMs are innately proangiogenic, exposure to tumor-derived ANG-2 stimulates these cells to exhibit a broader, tumor-promoting phenotype. As such, the ANG-2-TEM axis may represent a new target for antiangiogenic cancer therapies.


Subject(s)
Angiopoietin-2/genetics , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/genetics , Macrophages/physiology , Monocytes/physiology , Receptor, TIE-2/biosynthesis , Angiopoietin-2/biosynthesis , Animals , Carcinoma, Lewis Lung/metabolism , Gene Expression Regulation, Neoplastic , Humans , Macrophages/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Transgenic , Monocytes/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptor, TIE-2/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
11.
Arterioscler Thromb Vasc Biol ; 30(7): 1325-32, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20431068

ABSTRACT

OBJECTIVE: Recent studies on cardiovascular progenitors have led to a new appreciation that paracrine factors may support the regeneration of damaged tissues. METHODS AND RESULTS: We used a shotgun proteomics strategy to compare the secretome of peripheral blood-derived smooth muscle progenitors (SPCs) with human aortic smooth muscle cells. The late-outgrowth SPCs produced fewer proteolytic enzymes and inflammatory cytokines and showed reduced invasive capacity. Similar to smooth muscle cells, SPCs secreted extracellular matrix. However, SPCs produced different matrix proteins, as evidenced by the truncation of proangiogenic domains in collagen alpha-1 (I) and increased production of periostin. Moreover, SPCs retained serum proteins, including proteoglycans, regulating collagen assembly; and pigment epithelium-derived factor, a potent inhibitor of angiogenesis. As a functional consequence, their conditioned medium was less angiogenic, as demonstrated by endothelial tube formation assays in vitro and implantation of Matrigel plugs into nude, severe combined immunodeficient mice (NOD/SCID). CONCLUSIONS: The present study represents an important conceptual development, suggesting that SPCs may contribute to extracellular matrix production.


Subject(s)
Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Proteomics , Stem Cells/metabolism , Animals , Aorta/metabolism , Blood Proteins/metabolism , Cells, Cultured , Chromatography, Reverse-Phase , Culture Media, Conditioned/metabolism , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Muscle, Smooth, Vascular/cytology , Paracrine Communication , Peptide Hydrolases/metabolism , Proteomics/methods , Reproducibility of Results , Tandem Mass Spectrometry
12.
Circulation ; 121(18): 2001-11, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20421519

ABSTRACT

BACKGROUND: Cell therapy is a promising option to improve functional recovery after ischemia. Several subsets of bone marrow-derived cells were shown to reduce infarct size and increase ejection fraction in experimental models of ischemia. The mechanisms underlying the functional improvement are diverse and have been shown to include paracrine effects of the injected cells, as well as a variable degree of differentiation to endothelial cells, pericytes, smooth muscle, and cardiac muscle. METHODS AND RESULTS: To elucidate the true nature of such plasticity and contribution to recovery, we engineered vectors that encoded inducible suicide genes under the control of endothelium (endothelial nitric oxide synthase)-, smooth muscle (SM22alpha)-, and cardiomyocyte (alpha-MHC)-specific promoters, thereby allowing selective depletion of the individual cell lineage acquired by the transplanted undifferentiated bone marrow-derived cells. Lentivirally delivered thymidine kinase, which converts the prodrug ganciclovir into a cytotoxic agent, was used to selectively eliminate cells 2 weeks after transplantation of bone marrow mononuclear cells in an acute myocardial infarction model. We demonstrate that elimination of transplanted endothelium-committed or SM22alpha-expressing cells, but not cardiac-committed cells, induced a significant deterioration of ejection fraction. Moreover, elimination of endothelial nitric oxide synthase-expressing cells 2 weeks after injection reduced capillary and arteriole density. CONCLUSIONS: This study demonstrates that elimination of bone marrow mononuclear cells reexpressing endothelial nitric oxide synthase particularly induced a deterioration of cardiac function, which indicates a functional contribution of the vascular cell fate decision of human bone marrow-derived mononuclear cells in vivo.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Animals , Aorta/cytology , Cell Lineage/physiology , Endothelial Cells/cytology , Epithelial Cells/cytology , Epithelial Cells/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Graft Survival , Green Fluorescent Proteins/genetics , Humans , Lentivirus/genetics , Male , Mice , Mice, Nude , Muscle, Smooth, Vascular/cytology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Rats , Recovery of Function/physiology , Stroke Volume/physiology , Thymidine Kinase/genetics , Transplantation, Heterologous , Umbilical Veins/cytology
13.
Blood ; 115(23): 4944-50, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20299512

ABSTRACT

MicroRNAs are endogenously expressed small noncoding RNAs that regulate gene expression on the posttranscriptional level. The miR-17-92 cluster (encoding miR-17, -18a, -19a/b, -20a, and miR-92a) is highly expressed in tumor cells and is up-regulated by ischemia. Whereas miR-92a was recently identified as negative regulator of angiogenesis, the specific functions of the other members of the cluster are less clear. Here we demonstrate that overexpression of miR-17, -18a, -19a, and -20a significantly inhibited 3-dimensional spheroid sprouting in vitro, whereas inhibition of miR-17, -18a, and -20a augmented endothelial cell sprout formation. Inhibition of miR-17 and miR-20a in vivo using antagomirs significantly increased the number of perfused vessels in Matrigel plugs, whereas antagomirs that specifically target miR-18a and miR-19a were less effective. However, systemic inhibition of miR-17/20 did not affect tumor angiogenesis. Further mechanistic studies showed that miR-17/20 targets several proangiogenic genes. Specifically, Janus kinase 1 was shown to be a direct target of miR-17. In summary, we show that miR-17/20 exhibit a cell-intrinsic antiangiogenic activity in endothelial cells. Inhibition of miR-17/20 specifically augmented neovascularization of Matrigel plugs but did not affect tumor angiogenesis indicating a context-dependent regulation of angiogenesis by miR-17/20 in vivo.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/biosynthesis , Multigene Family , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Animals , Cell Line, Tumor , Endothelial Cells/pathology , Humans , Mice , MicroRNAs/genetics , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
14.
Circ Res ; 106(7): 1290-302, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20185800

ABSTRACT

RATIONALE: Complementation of pluripotency genes may improve adult stem cell functions. OBJECTIVES: Here we show that clonally expandable, telomerase expressing progenitor cells can be isolated from peripheral blood of children. The surface marker profile of the clonally expanded cells is distinct from hematopoietic or mesenchymal stromal cells, and resembles that of embryonic multipotent mesoangioblasts. Cell numbers and proliferative capacity correlated with donor age. Isolated circulating mesoangioblasts (cMABs) express the pluripotency markers Klf4, c-Myc, as well as low levels of Oct3/4, but lack Sox2. Therefore, we tested whether overexpression of Sox2 enhances pluripotency and facilitates differentiation of cMABs in cardiovascular lineages. METHODS AND RESULTS: Lentiviral transduction of Sox2 (Sox-MABs) enhanced the capacity of cMABs to differentiate into endothelial cells and cardiomyocytes in vitro. Furthermore, the number of smooth muscle actin positive cells was higher in Sox-MABs. In addition, pluripotency of Sox-MABs was shown by demonstrating the generation of endodermal and ectodermal progenies. To test whether Sox-MABs may exhibit improved therapeutic potential, we injected Sox-MABs into nude mice after acute myocardial infarction. Four weeks after cell therapy with Sox-MABs, cardiac function was significantly improved compared to mice treated with control cMABs. Furthermore, cell therapy with Sox-MABs resulted in increased number of differentiated cardiomyocytes, endothelial cells, and smooth muscle cells in vivo. CONCLUSIONS: The complementation of Sox2 in Oct3/4-, Klf4-, and c-Myc-expressing cMABs enhanced the differentiation into all 3 cardiovascular lineages and improved the functional recovery after acute myocardial infarction.


Subject(s)
Ischemia/surgery , Leukocytes, Mononuclear/transplantation , Muscle, Skeletal/blood supply , Myocardial Infarction/surgery , Peripheral Blood Stem Cell Transplantation , Pluripotent Stem Cells/transplantation , Regeneration , SOXB1 Transcription Factors/metabolism , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Female , Gene Expression Regulation, Developmental , Genetic Vectors/genetics , Hindlimb , Humans , Infant , Infant, Newborn , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Lentivirus/genetics , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Nude , Middle Aged , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/transplantation , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/transplantation , Neovascularization, Physiologic , Octamer Transcription Factor-3/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , Time Factors , Transduction, Genetic , Young Adult
15.
Science ; 324(5935): 1710-3, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19460962

ABSTRACT

MicroRNAs (miRs) are small noncoding RNAs that regulate gene expression by binding to target messenger RNAs (mRNAs), leading to translational repression or degradation. Here, we show that the miR-17approximately92 cluster is highly expressed in human endothelial cells and that miR-92a, a component of this cluster, controls the growth of new blood vessels (angiogenesis). Forced overexpression of miR-92a in endothelial cells blocked angiogenesis in vitro and in vivo. In mouse models of limb ischemia and myocardial infarction, systemic administration of an antagomir designed to inhibit miR-92a led to enhanced blood vessel growth and functional recovery of damaged tissue. MiR-92a appears to target mRNAs corresponding to several proangiogenic proteins, including the integrin subunit alpha5. Thus, miR-92a may serve as a valuable therapeutic target in the setting of ischemic disease.


Subject(s)
Endothelial Cells/metabolism , Ischemia/physiopathology , MicroRNAs/metabolism , Myocardial Infarction/physiopathology , Neovascularization, Physiologic , Animals , Antagomirs , Apoptosis/drug effects , Down-Regulation , Gene Expression Profiling , Hindlimb/blood supply , Humans , Integrin alpha5/genetics , Integrin alpha5/metabolism , Ischemia/drug therapy , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Muscle, Skeletal/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Oligoribonucleotides/pharmacology , Oligoribonucleotides/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regional Blood Flow , Up-Regulation , Ventricular Function, Left/drug effects , Zebrafish
16.
Blood ; 114(3): 723-32, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19369228

ABSTRACT

The concept of endothelial progenitor cells (EPCs) has attracted considerable interest in cardiovascular research, but despite a decade of research there are still no specific markers for EPCs and results from clinical trials remain controversial. Using liquid chromatography-tandem mass spectrometry, we analyzed the protein composition of microparticles (MPs) originating from the cell surface of EPC cultures. Our data revealed that the conventional methods for isolating mononuclear cells lead to a contamination with platelet proteins. Notably, platelets readily disintegrate into platelet MPs. These platelet MPs are taken up by the mononuclear cell population, which acquires "endothelial" characteristics (CD31, von Willebrand factor [VWF], lectin-binding), and angiogenic properties. In a large population-based study (n = 526), platelets emerged as a positive predictor for the number of colony-forming units and early outgrowth EPCs. Our study provides the first evidence that the cell type consistent with current definitions of an EPC phenotype may arise from an uptake of platelet MPs by mononuclear cells resulting in a gross misinterpretation of their cellular progeny. These findings demonstrate the advantage of using an unbiased proteomic approach to assess cellular phenotypes and advise caution in attributing the benefits in clinical trials using unselected bone marrow mononuclear cells (BMCs) to stem cell-mediated repair.


Subject(s)
Blood Platelets/cytology , Cell-Derived Microparticles/metabolism , Endothelial Cells/cytology , Leukocytes, Mononuclear/metabolism , Stem Cells/cytology , Bone Marrow Cells , Cells, Cultured , Chromatography, Liquid , Diagnostic Errors , Humans , Leukocytes, Mononuclear/cytology , Proteomics/methods , Research Design , Tandem Mass Spectrometry
17.
Blood ; 113(22): 5669-79, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19351956

ABSTRACT

Class IIa histone deacetylases (HDACs) are signal-responsive regulators of gene expression involved in vascular homeostasis. To investigate the differential role of class IIa HDACs for the regulation of angiogenesis, we used siRNA to specifically suppress the individual HDAC isoenzymes. Silencing of HDAC5 exhibited a unique pro-angiogenic effect evidenced by increased endothelial cell migration, sprouting, and tube formation. Consistently, overexpression of HDAC5 decreased sprout formation, indicating that HDAC5 is a negative regulator of angiogenesis. The antiangiogenic activity of HDAC5 was independent of myocyte enhancer factor-2 binding and its deacetylase activity but required a nuclear localization indicating that HDAC5 might affect the transcriptional regulation of gene expression. To identify putative HDAC5 targets, we performed microarray expression analysis. Silencing of HDAC5 increased the expression of fibroblast growth factor 2 (FGF2) and angiogenic guidance factors, including Slit2. Antagonization of FGF2 or Slit2 reduced sprout induction in response to HDAC5 siRNA. Chromatin immunoprecipitation assays demonstrate that HDAC5 binds to the promoter of FGF2 and Slit2. In summary, HDAC5 represses angiogenic genes, such as FGF2 and Slit2, which causally contribute to capillary-like sprouting of endothelial cells. The derepression of angiogenic genes by HDAC5 inactivation may provide a useful therapeutic target for induction of angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Histone Deacetylases/physiology , Neovascularization, Physiologic/genetics , Angiogenesis Inhibitors/antagonists & inhibitors , Angiogenesis Inhibitors/physiology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/physiology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/physiology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Models, Biological , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/physiology
18.
Arterioscler Thromb Vasc Biol ; 29(4): 571-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19122169

ABSTRACT

OBJECTIVE: Endothelial progenitor cells (EPCs) comprise a heterogeneous population of cells, which improve therapeutic neovascularization after ischemia. The neovascularization-promoting potential of progenitor cells depends on survival and retention of the infused cells to the tissue. Caspases mediate apoptosis but are also involved in other critical biological processes. Therefore, we aimed to address the role of caspases in proangiogenic cells. METHODS AND RESULTS: The caspase-8 inhibitor zIETD abrogated the ex vivo formation of EPCs, inhibited EPC adhesion and migration, and reduced their capacity to improve neovascularization in vivo. Consistently, cells isolated from caspase-8-deficient mice exhibited a reduced capacity for enhancing neovascularization when transplanted into mice after hindlimb ischemia. Because inhibition of Caspase-8 reduced the adhesion and homing functions of EPCs, we further determined the surface expression of integrins and receptors involved in cell recruitment to ischemic tissues. Pharmacological inhibition of caspase-8 and genetic depletion of caspase-8 reduced the expression of the fibronectin receptor subunits alpha5 and beta1 and the SDF-1 receptor CXCR4. Moreover, we identified the E3 ubiquitin ligase Cbl-b, which negatively regulates integrin and receptor-mediated signaling, as a potential Caspase-8 substrate. CONCLUSIONS: In summary, our data demonstrate a novel apoptosis-unrelated role of caspase-8 in proangiogenic cells.


Subject(s)
Caspase 8/metabolism , Endothelial Cells/enzymology , Ischemia/enzymology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Stem Cells/enzymology , Adaptor Proteins, Signal Transducing/metabolism , Alstrom Syndrome , Animals , Caspase 8/genetics , Caspase Inhibitors , Cell Adhesion , Cell Movement , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/transplantation , Hindlimb , Humans , Integrin alphaV/metabolism , Integrin beta1/metabolism , Ischemia/physiopathology , Ischemia/surgery , Mice , Mice, Knockout , Mice, Nude , Neovascularization, Physiologic/drug effects , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-cbl/metabolism , RNA, Messenger/metabolism , Receptors, CXCR4/metabolism , Receptors, Fibronectin/metabolism , Stem Cell Transplantation , Stem Cells/drug effects
19.
Circ Res ; 103(11): 1327-34, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18927463

ABSTRACT

Circulating blood-derived vasculogenic cells improve neovascularization of ischemic tissue by a broad repertoire of potential therapeutic actions. Whereas initial studies documented that the cells incorporate and differentiate to cardiovascular cells, other studies suggested that short-time paracrine mechanisms mediate the beneficial effects. The question remains to what extent a physical incorporation is contributing to the beneficial effects of cell therapy. By using the inducible suicide gene thymidine kinase to deplete transplanted cells, we determined the contribution of physical incorporation in 3 animal models. After acute myocardial infarction, depletion of cells 14 days after infusion resulted in a reduction of capillary density and a substantial deterioration of heart function. Likewise, neovascularization of Matrigel plugs and ischemic limbs was significantly suppressed when infused cells were depleted 7 days after infusion. Induction of cell death in the previously transplanted cells reduced perfusion and led to vascular leakage as evidenced by Evans blue extravasation. These results indicate that physical incorporation and persistence of cells contribute to cell-mediated improvement of neovascularization and cardiac function. Long-term paracrine activities and/or cell intrinsic mechanisms may have contributed to the maintenance of functional improvement.


Subject(s)
Heart/physiopathology , Myocardial Ischemia/therapy , Neovascularization, Physiologic/physiology , Stem Cell Transplantation/methods , Animals , Capillaries/physiopathology , Cell Culture Techniques , Cell- and Tissue-Based Therapy/methods , Coronary Vessels/pathology , Female , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Humans , Lentivirus , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/physiology , Magnetic Resonance Imaging , Mice , Mice, Nude , Myocardial Ischemia/physiopathology
20.
Circ Res ; 103(8): 796-803, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18776043

ABSTRACT

Therapeutic mobilization of vasculogenic progenitor cells is a novel strategy to enhance neovascularization for tissue repair. Prototypical mobilizing agents such as granulocyte colony-stimulating factor mobilize vasculogenic progenitor cells from the bone marrow concomitantly with inflammatory cells. In the bone marrow, mobilization is regulated in the stem cell niche, in which endosteal cells such as osteoblasts and osteoclasts play a key role. Because Wnt signaling regulates endosteal cells, we examined whether the Wnt signaling antagonist Dickkopf (Dkk)-1 is involved in the mobilization of vasculogenic progenitor cells. Using TOP-GAL transgenic mice to determine activation of beta-catenin, we demonstrate that Dkk-1 regulates endosteal cells in the bone marrow stem cell niche and subsequently mobilizes vasculogenic and hematopoietic progenitors cells without concomitant mobilization of inflammatory neutrophils. The mobilization of vasculogenic progenitors required the presence of functionally active osteoclasts, as demonstrated in PTPepsilon-deficient mice with defective osteoclast function. Mechanistically, Dkk-1 induced the osteoclast differentiation factor RANKL, which subsequently stimulated the release of the major bone-resorbing protease cathepsin K. Eventually, the Dkk-1-induced mobilization of bone marrow-derived vasculogenic progenitors enhanced neovascularization in Matrigel plugs. Thus, these data show that Dkk-1 is a mobilizer of vasculogenic progenitors but not of inflammatory cells, which could be of great clinical importance to enhance regenerative cell therapy.


Subject(s)
Bone Marrow Cells/metabolism , Cell Movement , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Physiologic , Osteoclasts/metabolism , Signal Transduction , Stem Cells/metabolism , Wnt Proteins/metabolism , Animals , Bone Marrow Cells/enzymology , Cathepsin K , Cathepsins/metabolism , Cell Differentiation , Cells, Cultured , Collagen , Drug Combinations , Female , Granulocyte Colony-Stimulating Factor/metabolism , Laminin , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteoclasts/enzymology , Proteoglycans , RANK Ligand/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Recombinant Proteins/metabolism , Stem Cells/enzymology , Time Factors , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL